You are on page 1of 109

Chapter 2

protozoan Infections

Intestinal Amebae
Pilarita T. Rivera, Windell L. Rivera, Juan Antonio A. Solon

S even species of amebae occur in humans.


These include the pathogenic Entamoeba
histolytica, and the commensals E. dispar, E.
reaction (PCR) restriction fragment length
polymorphism (RFLP), and typing with
monoclonal antibodies, these three species are
moshkovskii, E. hartmanni, E. coli, Endolimax now differentiated. E. hartmanni, formerly
nana, and Iodamoeba butschlii. Entamoeba referred to as “small race” of E. histolytica, is
polecki is an intestinal ameba of pigs and differentiated primarily on the basis of size.
monkeys that has been occasionally detected
Parasite Biology
in humans, and is a probable cause of diarrhea.
They are mainly differentiated on the basis of Entamoeba histolytica is a pseudopod-
structure and size. Trophozoites divide by binary forming non-flagellated protozoan parasite. It
fission. Most cyst-forming amebae go through is the most invasive of the Entamoeba parasites
nuclear division, and then divide again after (which includes E. dispar, E. moshkovskii, E.
excystation in a new host. hartmanni, E. polecki, E. coli, and E. gingivalis),
and the only member of the family to cause colitis
Entamoeba histolytica
and liver abscess. The life cycle of E. histolytica
Entamoeba histolytica is currently classified consists of two stages: an infective cyst (Plate
within the subphylum Sarcodina, superclass 2.1) and an invasive trophozoite form. No host
Rhizopoda, class Lobosea, order Amoebida, other than humans is implicated in the life cycle,
family Entamoebidae, and genus Entamoeba.
The members of this genus are characterized
by having a vesicular nucleus, a centrally (or near
central) located small karyosome, and varying
numbers of chromatin granules adhering to
the nuclear membrane. These nuclear and
other morphologic differences distinguish
the species of Entamoeba except E. histolytica,
E. dispar, and E. moshkovskii (previously
known as the Laredo strain). The three said
species are morphologically identical and of
the same size. It was only recently that this
E. histolytica species complex was resolved. Plate 2.1. Entamoeba histolytica cyst (Courtesy
Through isoenzyme analysis polymerase chain of the Department of Parasitology, UP-CPH)

20
Chapter 2: protozoan Infections 21

although natural infection of primates has been Infection with E. histolytica occurs when cysts
reported. The quadrinucleate cyst is resistant to are ingested from fecally-contaminated material
gastric acidity and desiccation, and can survive (Figure 2.1). Other modes of transmission
in a moist environment for several weeks. include venereal transmission through fecal-oral

Figure 2.1. Life cycle of Entamoeba histolytica


(Accessed from www.dpd.cdc.gov/dpdx)
22 MedICal parasItology In the phIlIppInes

contact or direct colonic inoculation through


contaminated enema equipment. Excystation
occurs in the small or large bowel, where a cyst
undergoes nuclear followed by cytoplasmic
division to form eight trophozoites. The E.
histolytica trophozoites are highly motile and
possess pseudopodia (Plate 2.2). They vary in
size from 12 to 60 µm in diameter (about 20 µm
in average). Microscopic examination of fully-
passed stool specimens reveals the characteristic
progressive and directional movement of
trophozoites, with pseudopodia as locomotory
Plate 2.2. Entamoeba histolytica trophozoite
organelles. The hyaline pseudopodium is (From World Health Organization. Bench Aids for
formed when the clear, glasslike ectoplasm, the Diagnosis of Intestinal Parasites.
or outer layer is extruded, and the granular Geneva: World Health Organization; 1994)
endoplasm flows into it. Ingested red blood
cells are observed as pale, greenish, refractile
bodies in the cytoplasm of the ameba. Cysts are
usually spherical, and the size may vary from 10
to 20 µm. They are characterized by a highly
refractile hyaline cyst wall, one to four nuclei,
and rod-shaped (or cigar-shaped) chromatoidal
bars. Trophozoites have the ability to colonize
and/or invade the large bowel, while cysts are
never found within invaded tissues. E. histolytica
trophozoites multiply by binary fission. They
encyst producing uninucleate cysts, which
then undergo two successive nuclear divisions Plate 2.3. Entamoeba histolytica quadrinucleate
to form the characteristic quadrinucleate cysts cyst (From World Health Organization. Bench Aids
(Plate 2.3). for the Diagnosis of Intestinal Parasites. Geneva:
E. histolytica is a eukaryotic organism but World Health Organization; 1994)
has several unusual features, including the lack
of organelles that morphologically resemble lack of glutathione metabolism, the use of
mitochondria. Because nuclear-encoded pyrophosphate instead of ATP at several steps
mitochondrial genes such as pyridine nucleotide in glycolysis, and the inability to synthesize
transhydrogenase and hsp60 are present, E. purine nucleotides de novo. Glucose is actively
histolytica, at one time may have contained transported into the cytoplasm, where the
mitochondria. There is no rough endoplasmic end products of carbohydrate metabolism are
reticulum or Golgi apparatus, although cell ethanol, carbon dioxide, and under aerobic
surface and secreted proteins contain signal conditions, acetate.
sequences, and tunicamycin inhibits protein Pathogenesis and Clinical Manifestations
glycosylation. Ribosomes form aggregated
crystalline arrays in the cytoplasm of the The proposed mechanisms for virulence
trophozoite. Some differences in biochemical are: production of enzymes or other cytotoxic
pathways from higher eukaryotes include the substances, contact-dependent cell killing,
Chapter 2: protozoan Infections 23

and cytophagocytosis. In vitro, amebic killing study involving 206 patients with probable
of target cultivated mammalian cells involve ALA as diagnosed by ultrasound, the two
receptor-mediated adherence of ameba to most frequent manifestations were fever in
target cells, amebic cytolysis of target cells, 77% and RUQ pain in 83%. Pain is either
and amebic phagocytosis of killed or viable localized in or referred to the right shoulder.
target cells. E. histolytica trophozoites adhere The liver is tender, especially in acute cases,
to the colonic mucosa through a galactose- and hepatomegaly is present in 50% of cases.
inhibitable adherence lectin (Gal lectin). Then, Chronic disease (>2 weeks duration) is found
the amebae kill mucosal cells by activation of in older patients and it involves wasting with
their caspase-3, leading to their apoptotic death significant weight loss rather than fever. Only
engulfment. 30% of ALA cases have concurrent diarrhea.
Recent studies have shown that susceptibility However, daily stool cultures revealed that 72%
of humans to E. histolytica infection is associated harbored trophozoites even in asymptomatic
with specific alleles of the HLA complex. infections. Mortality in uncomplicated ALA is
Majority of cases present as asymptomatic less than 1%.
infections with cysts being passed out in The onset of amebic colitis may be sudden
the stools (cyst carrier state). The recent after an incubation period of 8 to 10 days, or
differentiation of E. dispar and E. histolytica after a long period of asymptomatic cyst carrier
by PCR has confirmed the high prevalence state. ALA may have all acute presentation of
of non-pathogenic E. dispar compared to the less than 2 weeks duration or a chronic one of
pathogenic E. histolytica. However, studies also more than 2 weeks duration. The recurrence
revealed that most E. histolytica infections in rate was found to be 0.29% in a five-year study
endemic communities are asymptomatic. of ALA in Mexico.
Amebic colitis clinically presents as gradual The most serious complication of amebic
onset of abdominal pain and diarrhea with or colitis is perforation and secondary bacterial
without blood and mucus in the stools. Fever peritonitis. Colonic perforation occurs in 60%
is not common and it occurs only in one third of fulminant colitis cases.
of patients. Although some patients may only In ALA, the most serious complications are
have intermittent diarrhea alternating with rupture into the pericardium with a mortality
constipation, children may develop fulminant rate of 70%, rupture into the pleura with
colitis with severe bloody diarrhea, fever, and mortality of 15 to 30%, and super infection.
abdominal pain. Intraperitoneal rupture, which occurs in 2 to
Ameboma occurs in less than 1% of 7.5% of cases, is the second most common
intestinal infections. It clinically presents as complication. However, it is not as serious as
a mass-like lesion with abdominal pain and colonic perforation because ALA is sterile.
a history of dysentery. It can be mistaken for Secondary amebic meningoencephalitis
carcinoma. Asymptomatic ameboma may also occurs in 1 to 2%, and it should be considered
occur. in cases of amebiasis with abnormal mental
Amebic liver abscess (ALA) is the most status. Renal involvement caused by extension
common extra-intestinal form of amebiasis. of ALA or retroperitoneal colonic perforation is
The cardinal manifestations of ALA are fever rare. Genital involvement is caused by fistulae
and right upper quadrant (RUQ) pain. Several from ALA and colitis or primary infection
studies have shown these two as the most through sexual transmission.
frequent complaints, particularly in acute Natural or innate immunity to E. histolytica
cases (<2 weeks duration). In a Philippine in the intestines involves mucin inhibition of
24 MedICal parasItology In the phIlIppInes

amebic attachment to the underlying mucosal Acute amebic colitis should be differentiated
cells. In the systemic circulation, the mechanism from bacillary dysentery of the following
is that of complement-mediated killing of etiology: Shigella, Salmonella, Campylobacter,
trophozoites. Acquired immunity primarily Yersinia, and enteroinvasive Escherichia coli
involves cell-mediated responses, although (Table 2.1). Although stools may be grossly
humoral responses may also contribute to bloody or heme-positive in both conditions,
anti-amebic immunity. Activated T-cells kill fever and significantly elevated leukocyte count
E. histolytica by: a) directly lysing trophozoites are less common in amebic colitis. Another
in a contact-dependent process; b) producing differential is inflammatory bowel disease.
cytokines which activate macrophages and other Amebic colitis should be ruled out before
effector cells (neutrophils and eosinophils); and steroid therapy for inflammatory bowel disease
c) providing helper effect for B-cell antibody is started because of the risk of developing toxic
production. In vitro studies using activated megacolon.
murine and human T-cells demonstrated The differential diagnoses of ALA include
significant killing of trophozoites in a contact- pyogenic liver abscess, tuberculosis of the liver,
dependent and antibody independent manner. and hepatic carcinoma. On the other hand,
Cytokine studies revealed that interferon (IFN) genital amebiasis should be differentiated
and interleukin (IL-2) may have a role in from carcinoma, tuberculosis, chancroid, and
activating macrophages for amebicidal activity. lymphogranuloma venereum.
More recent studies demonstrated that activated
macrophages produce nitric oxide (NO) which Table 2.1. Comparison of bacillary and amebic
was lethal to trophozoites. Tumor necrosis factor dysentery
(TNF) was shown to stimulate NO production.
Bacillary Dysentery Amebic Dysentery
Although it is known that antibodies are
May be epidemic Seldom epidemic
produced against amebic antigens, there has
Acute onset Gradual onset
been no direct evidence of T-cell help for
Prodromal fever and No prodromal features
B-cells. Studies have revealed that the principal malaise common
antibody-dependent cell cytotoxicity (ADCC) Vomiting common No vomiting
did not work against amebae. Antibodies which Patient prostrate Patient usually ambulant
were detected by seroepidemiologic studies and
Watery, bloody diarrhea Bloody diarrhea
secretory IgA isolated in the gut may merely
Odorless stool Fishy odor stool
be an indicator of current or recent invasive
Stool microscopy:
amebiasis. numerous bacilli, pus
Amebic modulation of host immune cells,
responses exists. For instance, infected human macrophages, red cells, Stool microscopy: few
no Charcot-Leyden bacilli, red cells,
subjects and animals have been shown to be in crystals trophozoites with
a state of immunosuppression during the acute ingested red blood
cells, Charcot-Leyden
stage of amebiasis. This state, characterized crystals
by T-cell hyporesponsiveness, suppressed Abdominal cramps Mild abdominal cramps
proliferation and cytokine production, depressed common and severe
delayed-type hypersensitivity (DTH), and Tenesmus common Tenesmus uncommon
macrophage suppression, is favorable for amebic Natural history: Natural history: lasts for
survival. It is the reversal of these modulatory spontaneous recovery weeks; dysentery
in a few days, weeks or returns after remission;
effects, which is the key in controlling amebiasis. more; no relapse infection persists for
years
Chapter 2: protozoan Infections 25

Diagnosis following morphologic structures are noted:


size of the cyst, number of nuclei, location and
The standard method of parasitologic
appearance of the karyosome, the characteristic
diagnosis is microscopic detection of the
appearance of chromatoid bodies, and presence
trophozoites and cysts in stool specimens.
of cytoplasmic structures such as glycogen
Ideally, a minimum of three stool specimens
vacuole. E. histolytica can, thus, be differentiated
collected on different days should be examined.
from the non-pathogenic species, E. hartmanni,
For detection of trophozoites, fresh stool
E. coli, E. nana, and Iodameba bütschlii. Stool
specimens should be examined within 30
culture using Robinson’s and Inoki medium is
minutes from defecation. Using the direct fecal
more sensitive than stool microscopy, but is not
smear (DFS) with saline solution alone, the
routinely available.
microscopist can observe trophozoite motility.
Differentiation between E. histolytica and
Unidirectional movement is characteristic
E. dispar is not possible by microscopy. This
of E. histolytica. Using saline and methylene
can only be done by PCR, enzyme-linked
blue, Entamoeba species will stain blue, thus,
immunosorbent assay (ELISA), and isoenzyme
differentiating them from white blood cells.
analysis. The last is primarily a research
Using saline and iodine, the nucleus and
technique. On the other hand, an ELISA-based
karyosome can be observed to differentiate E.
assay for stool is now commercially available
histolytica from the non-pathogenic amebae
and studies have demonstrated a sensitivity of
(E. hartmanni, E. coli, Endolimax nana).
80% and specificity of 99%. The use of PCR
The detection of E. histolytica trophozoites
is limited by the requirement of sophisticated
with ingested red blood cells is diagnostic of
equipment. A Philippine study (n=497 stool
amebiasis. Charcot-Leyden crystals (Plate 2.4)
samples) looked into the reliability of stool
can also be seen in the stool.
ELISA with PCR as gold standard (Plate 2.5).
Sensitivity and specificity were 91% and 97%,
respectively.
Detection of antibodies in the serum is
still the key in the diagnosis of ALA. It must
be noted that in ALA, microscopic detection
cannot be done because aspiration is an invasive
procedure, and trophozoites are missed because
they are located in the periphery of the abscess.

Plate 2.4. Charcot-Leyden crystal observed


in stool specimen of a patient suffering from
amebiasis (Courtesy of the Department of
Parasitology, UP-CPH)

Concentration methods such as Formalin


Ether/Ethyl Acetate Concentration Test
Plate 2.5. Agarose gel showing the 100bp PCR
(FECT) and Merthiolate Iodine Formalin products of Entamoeba histolytica-positive
Concentration Test (MIFC) are more sensitive stool specimens (lanes 2-15)
than the DFS for detection of cysts. The (Courtesy of Dr. Windell Rivera)
26 MedICal parasItology In the phIlIppInes

To date, serological tests for amebic disease Treatment and Prognosis


include indirect hemagglutination (IHAT),
The treatment of amebiasis has two
counter immunoelectrophoresis (CIE), agar gel
objectives: a) to cure invasive disease at both
diffusion (AGD), indirect fluorescent antibody
intestinal and extraintestinal sites; and b) to
test (IFAT), and ELISA. The IHAT can detect
eliminate the passage of cysts from the intestinal
antibodies of a past infection even as long as 10
lumen. Metronidazole is the drug of choice
years ago. In contrast, the antibodies detected by
for the treatment of invasive amebiasis. Other
ELISA, AGD, and CIE are of short duration,
5-nitroimidazole derivatives such as tinidazole
lasting for a few months. Antibodies have
and secnidazole are also effective. Diloxanide
been demonstrated in asymptomatic intestinal
furoate is the drug of choice for asymptomatic
infections so that serology can be used in the
cyst passers. It is also given after a course of
monitoring of a cyst carrier.
metronidazole for invasive amebiasis.
Ultrasound, computerized tomography
Percutaneous drainage of liver abscess is
(CT scan), and magnetic resonance imaging
indicated for patients who do not respond
(MRI) are non-invasive and sensitive methods
to metronidazole and who need prompt
in early detection of ALA. Ultrasound (Plate
symptomatic relief of severe pain. It is also done
2.6) typically shows a round or oval hypoechoic
for those who have left lobe abscess that may
area with wall echoes. In 80% of cases, this
rupture into the pericardium, large abscesses in
finding is seen in the right lobe of the liver.
danger of rupture, and multiple abscesses with
Multiple lesions occur in 50% of acute cases,
a probable associated pyogenic etiology.
and aspiration may be required to differentiate
amebic from pyogenic abscess. Using serological Epidemiology
methods (IHAT and IFAT) as gold standard, a
For a long time, the species-complex
Philippine study has shown that the sensitivity
referred to as E. histolytica was believed to
and specificity of ultrasound were 95% and
infect 500 million people, or 10% of the
40%, respectively. However, as the results of
world’s population. However, with the recent
the study still revealed some limitations in the
redescription into three different species: the
use of ultrasound in the diagnosis of ALA,
pathogenic E. histolytica, and the commensals, E.
additional diagnostic ultrasound findings have
dispar and E. moshkovskii, the true prevalence of
yet to be identified.
amebiasis is approximately 1 to 5% worldwide.
There are 50 million E. histolytica infection
cases, and 40,000 to 100,000 deaths due to
amebiasis in the world per year. Thus, amebiasis
is the third most important parasitic disease,
after malaria and schistosomiasis, and second
to malaria as the top cause of mortality among
parasitic protozoans.
Humans are the major reservoirs of
infection with E. histolytica. Ingestion of food
and drink contaminated with E. histolytica
cysts from human feces, and direct fecal-
oral contact are the most common means of
Plate 2.6. Ultrasound showing a solitary infection. Amebic infection is prevalent in the
hypoechoic mass at the right lobe of the liver Indian subcontinent, Africa, East Asia, and
suggesting ALA (Courtesy of Dr. Pilarita Rivera) South and Central America. In developing
Chapter 2: protozoan Infections 27

countries, prevalence depends on the level of cases should be done. Food handlers should be
sanitation, crowding, socio-economic status, screened for cyst carriage, and asymptomatic
cultural habits, and age. In developed countries, cyst carriers should be treated.
infection is usually caused by E. dispar, and Vaccines can be a cost-effective and
is prevalent in certain groups: immigrants, potent strategy for amebiasis prevention
travelers from endemic countries, homosexual and eradication. Unlike in other protozoan
males (men having sex with men), HIV patients, infections, amebic vaccine development has
and institutionalized people. fewer problems. The ameba life cycle is simple,
A microscopic study of diarrheic stools in and no intermediate hosts are involved. Amebae
Australia (n=5,921) revealed 177 (3%) positive are extracellularly located, and do not undergo
samples. PCR detected 5 E. histolytica, 63 E. antigenic variation. All these characteristics are
dispar, and 55 E. moshkovskii infections. The supportive of an achievable amebic vaccine.
latter two species, which are both commensals, Studies have also demonstrated the
are 10 times more prevalent than E. histolytica. acquisition of protective immunity to amebae,
A stool survey done in Iran (n=16,592) showed particularly that of mucosal immune response.
226 positive samples. Only 101 isolates were Trials with recombinant amebic antigens as
successfully cultured in Robinson’s medium. vaccines have proven to be more advantageous
Of these isolates, 93 (92.1%) were E. dispar, than inactivated/attenuated amebae. The
and only 8 (7.9%) were E. histolytica or mixed candidate vaccine molecules which have been
infections by PCR- RFLP. most intensely studied are the serine-rich E.
A field study in Northern Philippines histolytica protein (SREHP), the adherence
(n=1,872) showed 137 (7.3%) E. dispar, and lectin (Gal/GalNAc lectin), and the 29 kDa
18 (0.96%) E. histolytica by PCR. A study in a cysteine-rich amebic antigen. However, most
mental institution (n=113) showed E. histolytica of these studies have utilized animal models
or E. dispar in 43 subjects (38.1%), while PCR and artificial infection during challenge.
detected 74 (65.5%) E. histolytica-positive Testing these candidate vaccines in humans
samples, and 6 (5.3%) E. dispar/E. histolytica and developing them as food-based vaccines
mixed samples. will be in the forefront of future directions of
amebiasis control.
Prevention and Control
References
The prevention and control of amebiasis
depends on integrated and community-based Ali IK, Clark CG, Petri WA Jr.. Molecular
efforts to improve environmental sanitation, epidemiology of amebiasis. Infect Genet
and to provide for sanitary disposal of human Evol. 2008;8(5):698–707.
feces, safe drinking water, and safe food. These Diamond LS, Clark CG. A redescription of
efforts become more sustainable through health Entamoeba histolytica Schaudinn, 1903
education and promotion. The proper use of (Emended Walker, 1911) separating it
latrines and practice of proper hygiene, such from Entamoeba dispar Brumpt, 1925. J
as washing of hands, should be emphasized. Eukaryot Microbiol. 1993;40:340–4.
In communities where potable water is not Farthing M, Cevallos A, Kelly P, Cook G.
available, drinking water should be boiled or Manson’s tropical disease. 20th ed. London:
filtered. Vegetables and fruits which are eaten WB Saunders Co. Ltd.; 1996. p. 1255–69.
raw should be thoroughly washed. The use Fotedar R, Stark D, Beebe N, Marriott D,
of night soil for fertilizer should be avoided. Ellis J, Harkness J. PCR Detection of
Prompt diagnosis and treatment of amebiasis Entamoeba histolytica, Entamoeba dispar,
28 MedICal parasItology In the phIlIppInes

and Entamoeba moshkovskii in stool samples histolytica and Entameba dispar in the
from Sydney, Australia. J Clin Microbiol. Northern Philippines as detected by the
2007;45(3):1035–7. polymerase chain reaction. Am J Trop Med
Garcia E, Tiu W. Immunodiagnosis of some Hyg. 1998;59(6):916–22.
parasitic infections in the Philippines. Rivera WL, Tachibana H, Kanbara H.
Taguig: National Academy of Science and Application of polymerase chain reaction
Technology; 1998. p. 23–8. (PCR) in the epidemiology of Entamoeba
Hira PR, Iqbal J, Al-Ali F, Philip R, Grover histolytica and Entamoeba dispar infections.
S, D’Almeida E, et al. Invasive amebiasis: Tokai J Exp Clin Med. 1999;23:413–5.
challenges in diagnosis in a nonendemic Rivera WL, Tachibana H, Silva-Tahat MR,
country (Kuwait). Am J Trop Med Hyg. Uemura H, Kanbara H. Differentiation of
2001;65(4):341–5. Entamoeba histolytica and E. dispar DNA
Hooshya H, Rezaian M, Kazemi B, Jeddi- from cysts present in stool specimens
Tehrani M, Solaymani-Mohammadi S. by polymerase chain reaction: its field
The distribution of Entamoeba histolytica application in the Philippines. Parasitol
and Entamoeba dispar in Northern, Res. 1996;82:585–9.
Central, and Southern Iran. Parasitol Res. Salazar N, Pasay C, Avenido A, Macapsir S,
2004;94:96–100. Lena M, Maguinsay V, et al. Detection
John DT, Petri WA. Markell and Voge’s medical of Entamoeba histolytica in routine stool
parasitology. 9th ed. St. Louis: Elsevier examination. Phil J Microbiol Infect Dis.
Saunders; 2006. p. 22–36. 1990; 19(23):57–66.
Petri WA Jr. Recent advances in amebiasis. Crit Stark DJ, Fotedar R, van Hal SJ. Prevalence of
Rev Clin Lab Sci. 1996;33(1):17–23. enteric protozoa in HIV-positive and HIV-
Ravdin JI. Amebiasis: series on tropical negative men who have sex with men from
medicine: science and practice. London: Sydney, Australia. Am J Trop Med Hyg.
Imperial College Press; 2000. p. 65–159. 2007;76:549–52.
Rivera PT, Rivera WL, Escueta AS, Villacorte Van Hal SJ, Stark DJ, Fotedar R, Marriott
EA, Limlingan ET, Lazaro CM, et al. D, Ellis JT, Harkness JL. Amoebiasis:
Prevalence of serologically positive amebic current status in Australia. Med J Aust.
liver abscess cases in the Philippines. Acta 2007;186(8):412–6.
Med Philipp. 2007;41(2):31–6. Walsh JA, Ravdin JI. Prevalence of Entamoeba
Rivera WL, Kanbara H. Detection of histolytica infection. Amebiasis. Human
Entamoeba dispar DNA in macaque feces Infection by Entamoeba histolytica. New
by polymerase chain reaction. Parasitol Res. York: Wiley; 1988. p. 93–105.
1999;85:493–5. World Health Organization. Entamoeba
Rivera WL, Tachibana H, Kanbara H. Field taxonomy. Bull World Health Organ.
Study on the distribution of Entameba 1997;75:291–2.
Chapter 2: protozoan Infections 29

Commensal Amebae
Pilarita T. Rivera, Vicente Y. Belizario, Jr., Juan Antonio A. Solon

T he presence of commensal amebae in the


stools of an individual is significant for two
reasons: (a) the amebae may be mistaken for the
(Figure 2.2). These amebae are non-invasive
and do not cause disease.
Reproduction is by binary fission of the
pathogenic Entamoeba histolytica; and (b) it is trophozoites. Encystation occurs as amebae pass
an indication of fecal contamination of food through the lower colon where colonic contents
or water. Accurate identification of commensal are more dehydrated.
amebae is therefore crucial.
Entamoeba dispar
Parasite Biology
Entamoeba moshkovskii
Commensal amebae must be differentiated Entamoeba dispar is morphologically
from pathogenic E. histolytica to avoid similar to E. histolytica, but their DNA and
unnecessary treatment of patients infected ribosomal RNA are different. The former’s
with non-pathogenic species. The three genera isoenzyme pattern is different from that of E.
of intestinal amebae can be differentiated histolytica.
through the morphological features of their Entamoeba moshkovskii isolates, although
nuclei. The genus Entamoeba has a spherical first detected in sewage, have been reported
nucleus with a distinct nuclear membrane in some areas, such as North America, Italy,
lined with chromatin granules and a small South Africa, Bangladesh, India, Iran, and
karyosome found near the center of the nucleus. Australia. It is a non-pathogenic species
Trophozoites usually have only one nucleus. The that is morphologically indistinguishable
genus Endolimax has a vesicular nucleus with from E. histolytica and E. dispar, but differs
a relatively large, irregularly-shaped karyosome from them biochemically and genetically. E.
anchored to the nucleus by achromatic fibrils. moshkovskii is also physiologically unique—it
The genus Iodamoeba is characterized by a large, being osmotolerant, able to grow at room
chromatin-rich karyosome surrounded by a temperature (25-30°C optimum), able to
layer of achromatic globules and anchored to survive at temperatures ranging from 0 to 41°C.
the nuclear membrane by achromatic fibrils. It has limited pathogenicity in experimental
All species have the following stages: trials in animals, but is non-pathogenic to
trophozoite, precyst, cyst, and metacystic humans. All human isolates have been found
trophozoite; with the exception of Entamoeba to belong to one group “ribodeme 2.”
gingivalis, which has no cyst stage, and does not
inhabit the intestines. Humans are infected by Entamoeba hartmanni
commensal intestinal amebae through ingestion The appearance of E. hartmanni is relatively
of viable cysts in food or water. Cysts pass similar to that of E. histolytica apart from its
through the acidic stomach and remain viable smaller size. Trophozoites of the former measure
because of protective cyst walls. Excystation from 3 to 12 µm in diameter (compared to E.
occurs in the alkaline environment of the histolytica measuring 12-60 µm). Mature cysts
lower small intestines. Metacystic trophozoites measure 4 to 10 µm, are quadrinucleated like
colonize the large intestines and live on the E. histolytica, and have rod-shaped chromatoid
mucus coat covering the intestinal mucosa material with rounded or squared ends. Unlike
30 MedICal parasItology In the phIlIppInes

Figure 2.2. Life cycle of commensal amebae


(Accessed from www.dpd.cdc.gov/dpdx)

E. histolytica, E. hartmanni does not ingest red by the following features: 1) a more vacuolated
blood cells. or granular endoplasm with bacteria and
debris, but no red blood cells; 2) a narrower,
Entamoeba coli
less-differentiated ectoplasm; 3) broader and
Entamoeba coli is cosmopolitan in blunter pseudopodia used more for feeding
distribution, and is considerably more common than locomotion; 4) more sluggish, undirected
than other human amebae. Trophozoites of E. movements; and 5) thicker, irregular peripheral
coli measure 15 to 50 µm in diameter. It can chromatin with a large, eccentric karyosome in
be differentiated from E. histolytica trophozoite the nucleus (Plate 2.7).
Chapter 2: protozoan Infections 31

Entamoeba chattoni, which is found in apes


and monkeys, is morphologically identical to
E. polecki. More recently, it has been detected
in eight human infections. Identification of E.
chattoni was done via isoenzyme analysis.
Entamoeba gingivalis

Entamoeba gingivalis can be found in


the mouth. The trophozoite measures 10 to
20 µm. It moves quickly, and has numerous
blunt pseudopodia. Food vacuoles that contain
cellular debris (mostly leukocytes, which is
characteristic of this species) and bacteria are
numerous. E. gingivalis lives on the surface of
Plate 2.7. Entamoeba coli trophozoite (Courtesy gum and teeth, in gum pockets, and sometimes
of Department of Parasitology, UP-CPH)
in the tonsillar crypts. They are abundant in
cases of oral disease. This species has no cyst
An E. coli cyst may be differentiated from stage. Transmission is most probably direct:
E. histolytica by: 1) its larger size (10 to 35 µm through kissing, droplet spray, or by sharing
in diameter), 2) more nuclei (eight versus four utensils.
in E. histolytica), 3) more granular cytoplasm,
and 4) splinter-like chromatoidal bodies. Iodine Endolimax nana
staining reveals dark-staining, perinuclear Endolimax nana occurs with the same
masses, which are actually glycogen. Its location, frequency as Entamoeba coli. Trophozoites
surrounding the nucleus, is more characteristic are small, with a diameter of 5 to 12 µm, and
of E. coli compared to E. histolytica. exhibit sluggish movement. They have blunt,
Entamoeba polecki hyaline pseudopodia, and the nucleus has a
large, irregular karyosome. Food vacuoles found
Entamoeba chattoni
in the cytoplasm may contain bacteria. Cysts
Entamoeba polecki is a parasite found in measure about the same size as trophozoites,
the intestines of pigs and monkeys. Rarely, it and are quadrinucleated when mature.
can infect humans, though a high prevalence Iodamoeba bütschlii
(19%) was reported in some parts of Papua
New Guinea (n=184 children). In these areas, The trophozoite averages 9 to 14 µm
both pig-to-human and human-to-human in diameter (ranging from 4-20 µm). It is
transmission may exist. Like E. coli, motility identified by its characteristic large, vesicular
of trophozoites of E. polecki is sluggish. A nucleus with a large, central karyosome,
small karyosome is centrally located in the surrounded by achromatic granules. There
nucleus. E. polecki can be distinguished from E. are no peripheral chromatin granules on the
histolytica in that the former’s cyst is consistently nuclear membrane. The cyst is about 9 to 10
uninucleated, and chromatoidal bars are µm in diameter (ranging from 6-16 µm), is
frequently angular or pointed. In stained fecal uninucleated, and has a large glycogen body
smears, the nuclear membrane and karyosome which stains dark brown with iodine (Plate 2.8).
are very prominent.
32 MedICal parasItology In the phIlIppInes

1998. A study on intestinal parasitic infections


among food service workers in a tertiary
hospital in Manila revealed that 20.3% were
infected with Endolimax nana and 13.6% with
Entamoeba coli. Another study of food handlers
in selected school canteens in Manila showed
infection rates of 22.8% for Endolimax nana,
17.9% for Entamoeba coli, and 0.8% each for
Entamoeba hartmanni and Iodamoeba bütschlii.
Prevention and Control

Contraction of the organism may be


prevented through proper disposal of human
Plate 2.8. Iodamoeba bütschlii cyst waste and good personal hygiene.
(Courtesy of the Department of Parasitology,
UP-CPH) References

Ali IK, Clark CG, Petri WA. Molecular


Diagnosis Epidemiology of Amebiasis. Infect Genet
Di a g n o s i s i s d o n e t h ro u g h s t o o l Evol. 2008;8(5):698–707.
examination. Formalin ether/ethyl acetate Antonio R. Conservative management of two
concentration technique (FECT) and iodine cases of hepato-pulmonary amebiasis. Bull
stain are useful to differentiate the species. For Quezon Inst. 1954;2:263–74.
E. gingivalis, a swab between the gums and teeth Avila MS, Garcia MR, Narcelles MV, Serra
is examined for trophozoites. Cysts are recovered FB, Tejida GM. Prevalence of intestinal
from formed stools, while trophozoites are helminth and protozoan infections among
recovered from watery or semi-formed stools. foodhandlers in selected school canteens
Trophozoites are best demonstrated by direct in Manila [undergraduate special study].
fecal smear. In recovering cysts, the use of 2003. Located at: College of Public Health
concentration techniques like FECT and zinc Library, University of the Philippines
sulfate flotation is useful. Manila.
Beaver PC, Jung RC, Cupp EW. Clinical
Treatment parasitology. 9th ed. Philadelphia: Lea &
No treatment is necessary because these Febiger; 1984.
amebae do not cause disease. Belding DL. Textbook of parasitology, 3rd
ed. New York: Appleton-Century Crofts;
Epidemiology 1965.
In single stool examinations of over Clark CA, Diamond LS. Pathogenicity,
30,000 Filipinos, the prevalence of Entamoeba virulence and Entamoeba histolytica.
coli was about 21%, Endolimax nana, about Parasitol Today. 1994;11(2):46–7.
9%, and Iodamoeba bütschlii, 1%. Intestinal Cross JH, Basaca-Sevilla V. Biomedical surveys
protozoan cysts were observed in 13.5% of in the Philippines. Manila (Philippines):
overseas Filipino workers (OFWs) screened by U.S. Naval Medical Research Unit No.
the Department of Parasitology, UP Manila in 2; 1984.
Chapter 2: protozoan Infections 33

Diamond LS, Clark CG. A redescription of Neva FA, Brown HW. Basic Clinical Parasitology.
Entamoeba histolytica Schaudinn, 1903 6th ed. Connecticut: Appleton & Lange;
(Emended Walker, 1911) separating it 1994.
from Entamoeba dispar Brumpt, 1925. J Phillips SC, Mildvan MD, William DC, Gelb
Eukaryot Microbiol. 1993;40(3):340–4. AM, White MC. Sexual transmission
Esparar DG, Belizario VY, Relos JR. Prevalence of enteric protozoa and helminths in a
of parasitic infection among food-handlers venereal disease clinic population. N Engl
in a dietary service of a tertiary hospital in J Med. 1981; 305(11):603–6.
Manila. Phil J Microbiol Infect Dis. 2004; Roberts LS, Janovy J. Foundations of
33(3):99–103. parasitology. 5th ed. Dubuque: Wm. C.
Imperato PJ. A historical overview of amebiasis. Brown Publishers; 1996.
Bull N Y Acad Med. 1981;57(3):175-87. Salazar NP, Pasay CJ, Avenido AO, Macapasir
John DT, Petri WA. Markell and Voge’s medical SR, Lena MJ, Maguinsay VM, et al.
parasitology. 9th ed. St. Louis: Elsevier Detection of Entamoeba histolytica in
Saunders; 2006. p. 36–48. routine stool examination. Phil J Microbiol
Mahmoud AA. Tropical and geographical Infect Dis. 1990;19(2):57–60.
medicine companion handbook. 2nd ed. Van Hal SJ, Stark DJ, Fotedar R, Marriott
Singapore: McGraw-Hill Book Co.; 1993. D, Ellis JT, Harkness JL. Amoebiasis:
Martinez-Palomo A, Espinosa-Castellano M. current status in Australia. Med J Aust.
Amoebiasis: new understanding and new 2007;186(8):412–6.
goals. Parasitol Today. 1998;14(1):1–4. World Health Organization. Amebiasis. Wkly
Epidemiol Rec. 1997;72(14):97–100.
34 MedICal parasItology In the phIlIppInes

Free-living Pathogenic Amebae


Edsel Maurice T. Salvana

Acanthamoeba spp.
Parasite Biology

A canthamoeba is a ubiquitous, free-living


ameba that is the etiologic agent of
Acanthamoeba keratitis (AK) and granulomatous
amebic encephalitis (GAE). Acanthamoeba is
characterized by an active trophozoite stage
with characteristic prominent “thorn-like”
appendages (acanthopodia); and a highly
resilient cyst stage into which it transforms when
environmental conditions are not favorable.
It is an aquatic organism that is found in a
myriad of natural and artificial environments,
and can survive even in contact lens cleaning
solutions. Motile trophozoites feed on gram- Plate 2.9. Acanthamoeba trophozoite exhibiting
negative bacteria, blue-green algae, or yeasts and characteristic acanthopodia (Accessed from
www.dpd.cdc.gov/dpdx)
reproduce by binary fission, but can also adapt
to feed on corneal epithelial cells and neurologic
tissue through phagocytosis and secretion of The presence of naturally-occurring
lytic enzymes. bacterial endosymbionts in Acanthamoeba
M o r p h o l o g i c a l l y, A c a n t h a m o e b a spp. has been reported. Although the presence
trophozoites exhibit a characteristic single of bacterial symbionts is widespread among
large nucleus with a centrally-located, densely small, free-living amebae, the significance
staining nucleolus; a large endosome; finely of this association is not known. Recently,
granulated cytoplasm; and a large contractile Acanthamoeba spp. have been implicated as
vacuole. Small, spiny filaments for locomotion possible reservoir hosts for medically important
known as acanthapodia are evident on phase- bacteria such as Legionella spp., mycobacteria,
contrast microscopy (Plate 2.9). and gram-negative bacilli such as E. coli.
Acanthamoeba has only two stages, cysts and Pathogenesis and Clinical Manifestations
trophozoites, in its life cycle. No flagellated stage
A. Acanthamoeba Keratitis
exists as part of the life cycle. The trophozoites
replicate by mitosis (nuclear membrane does Acanthamoeba was first described as an
not remain intact). The trophozoites are opportunistic ocular surface pathogen causing
the infective stage, although both cysts and keratitis in 1974. AK is associated with the
trophozoites gain entry into the body through use of improperly disinfected soft contact
various means. Entry can occur through the lenses, particularly those which are rinsed with
eye, the nasal passages to the lower respiratory tap water or contaminated lens solution. An
tract, or ulcerated or broken skin (Figure 2.3). immunocompromised state contributes to
Chapter 2: protozoan Infections 35

Figure 2.3. Life cycle of Acanthamoeba spp.


(Accessed from www.dpd.cdc.gov/dpdx)

increased susceptibility to infection, and may fluorescence microscopy. GAE usually occurs
lead to disseminated disease in the lungs and in immunocompromised hosts including
brain (GAE). the chronically ill and debilitated, and
Symptoms of AK include severe ocular those on immunosuppressive agents such as
pain and blurring of vision. Corneal ulceration chemotherapy and anti-rejection medications.
with progressive corneal infiltration may occur. The acquired immune deficiency syndrome
Primary amebic infection or secondary bacterial (AIDS) epidemic in the 1980’s dramatically
infection may lead to hypopyon formation. increased the numbers of person with GAE,
Progression of infection may cause scleritis and but these numbers have since fallen with the
iritis, and may ultimately lead to vision loss. advent of highly effective antiretroviral therapy.
Major differentials which need to be ruled out Signs and symptoms of GAE are generally
include fungal and herpetic keratitis. related to destruction of brain tissue and the
associated meningeal irritation. Systemic
B. Granulomatous Amebic Encephalitis
manifestations early in the course include fever,
Acanthamoeba was documented as the malaise, and anorexia. Neurologic symptoms
causative agent of human GAE by Stamm in may include increased sleeping time, severe
1972. Amebae were demonstrated in brain headache, mental status changes, epilepsy, and
sections of a GAE patient using indirect coma. Neurologic findings depending on the
36 MedICal parasItology In the phIlIppInes

location of the lesions include hemiparesis, Diagnosis of GAE is usually made post-
blurring of vision, diplopia, cranial nerve mortem in most cases. The rarity of the
deficits, ataxia, and increased intracranial disease and unfamiliarity of most physicians
pressure. with the pathogen contribute to frequently
Entry of Acanthamoeba into the central missed diagnosis. Signs and symptoms of
nervous system is still incompletely understood. disease are usually attributed to more common
From a primary site of infection in the differentials. Moreover, recovery of ameba from
skin or lungs, the likely route of invasion is cerebrospinal fluid is exceedingly rare, and
hematogenous. Direct infection through the imaging results are generally nonspecific.
olfactory valves has also been proposed, but Immunocompromised patients such
not conclusively demonstrated. Recent reviews as those with AIDS are at the highest risk
have focused on blood-borne invasion, with for acquiring GAE. While opportunistic
a combination of host factors, elucidation of infections of the central nervous system such
serine proteases, and parasite adhesion using as Cryptococcus meningitis and toxoplasmosis
a mannose-binding protein all contributing to are much more common than GAE, the lack of
brain endothelial cell damage and subsequent response despite appropriate treatment should
breakdown of the blood-brain barrier. prompt a more thorough evaluation for more
Gross examination of neural tissue post- esoteric organisms.
mortem reveals cerebral hemispheres that are Specific diagnosis depends on demonstrating
edematous and soft, with areas of hemorrhage the trophozoites or cysts in tissues using
and focal abscesses. The most affected areas histopathologic stains and microscopy. The
of the brain are the posterior fossa structures, organisms can rarely be demonstrated in the
thalamus, and the brainstem. In the affected cerebrospinal fluid and can be cultured for
areas, the leptomeninges are opaque and exhibit further studies.
purulent exudates and vascular congestion.
Treatment
The incubation period from initial
inoculation is approximately 10 days, with a Medical treatment of AK has been met
subacute and chronic clinical course of infection with increasing success in recent years. While
that lasts for several weeks to several months. historically, only surgical excision of the infected
The clinical manifestations of disease include cornea with subsequent corneal transplantation
decreased sensorium, altered mental status, was curative, early recognition of AK coupled
meningitis, and neurologic deficits. The natural with aggressive combination anti-amebic
course of the disease eventually results in coma agents can preclude the need for extensive
and death. surgery. D’Aversa and his colleagues have
achieved acceptable results with clotrimazole
Diagnosis
combined with pentamidine, isethionate,
Acanthamoeba keratitis is diagnosed by and neosporin. Other agents that have been
epithelial biopsy or corneal scrapings for used include polyhexamethylene biguanide,
recoverable ameba with characteristic staining propamidine, dibromopropamidine isethionate,
patterns on histologic analysis. Amebae have neomycin, paromomycin, polymyxin B,
also been isolated from the contact lens and lens ketoconazole, miconazole, and itraconazole.
solution of patients. Species-specific identification Topical corticosteroids should be avoided, as
can be made from culture and molecular analysis this retards the immune response. Advanced
through PCR. Known species that have caused AK usually requires debridement, but complete
AK include A. castellani, A. culbertsoni, A. excision of the cornea can be avoided if the
hutchetti, A. polyphaga, and A. rhysoides. infection is confined to more superficial areas.
Chapter 2: protozoan Infections 37

Deep lamellar keratectomy is the procedure of of the risk of infection, and physicians treating
choice. these patients should maintain a high index
Clinically apparent neurologic disease in of suspicion in the presence of compatible
GAE usually heralds a fatal outcome within signs and symptoms of infection which do not
3 to 40 days. A few patients have shown good respond to conventional antimicrobial therapy.
responses to combinations of amphotericin
References
B, pentamidine isethionate, sulfadiazine,
flucytosine, fluconazole or itraconazole. D’Aversa G, Stern GA, Driebe WT Jr. Diagnosis
One liver transplant patient survived after and successful medical treatment of
decompressive frontal lobectomy and treatment Acanthamoeba keratitis. Arch Ophthalmol.
with amphotericin, cotrimoxazole, and 1995;113(9):1120–3.
rifampin. Poor prognostic factors include severe De Jonckheere JF. Ecology of Acanthamoeba.
immunosuppression and advanced disease. Rev Infect Dis. 1991;13(Suppl 5):S3857.
Enriquez GL, Lagmay J, Natividad FF, Matias
Epidemiology
GA. Pathogenicity of two human isolates of
Acanthamoeba spp. have a protean Acanthamoeba keratitis in mice. Proc. IXth
distribution, having been isolated from a International Congress of Protozoology;
multitude of natural and artificial aquatic 1993. Berlin, Germany.
environments including fresh and salt water, Fung KT, Dhillon AP, McLaughlin JE, Lucas
sewage, hospital equipment, and contact lenses SB, Davidson B, Rolles K, et al. Cure of
and lens solution. Acanthamoeba cerebral abscess in a liver
De Jonckheere first diagnosed Acanthamoeba transplant patient. Liver Transpl. 2008;
GAE in a living patient in 1991. Previously, 14(3):308–12.
diagnosis of GAE was post-mortem. AK was Khan NA. Acanthamoeba and the blood-
recognized earlier in the 1970s and has been brain barrier: the breakthrough. J Med
reported in the United States, Europe, South Microbiol. 2008;57:1051–7.
America, and Asia. The first case of AK was Matias R, Schottelius J, Raddatz CF, Michel R.
recognized in the Philippines in the 1990s Species identification and characterization
from a patient from the Philippine General of an Acanthamoeba strain from human
Hospital, and samples obtained from the patient cornea. Parasitol Res. 1991;77(6):469–74.
was shown to cause GAE in mice. Multiple Salvana EM, Matias RR. Histopathology of
environmental isolates have likewise been well- mouse brain infected with Acanthamoeba
characterized from all over the Philippines, isolate IB-17 [undergraduate thesis].
including a few containing endosymbionts. Quezon City, Philippines: University of
the Philippines Diliman; 1996.
Prevention and Control
Visvesvara GS, Moura H, Schuster FL.
The ubiquitious nature of Acanthamoeba Pathogenic and opportunistic free-living
spp. makes exposure unavoidable. A robust amoebae: Acanthamoeba spp., Balamuthia
immune system is able to prevent infection, mandrillaris, Naegleria fowleri, and Sappinia
except in relatively immunocompromised diploidea. FEMS Immunol Med Microbiol.
sites such as the cornea. Meticulous contact 2007;50:1–26.
lens hygiene is essential in avoiding infection, Yagita K, Matias RR, Yasuda T, Natividad FF,
and rinsing contact lenses in tap water should Enriquez GL, Endo T. Acanthamoeba sp.
be avoided. Prolonged heating and boiling from the Philippines: electron microscopy
kill amebic trophozoites and cyst forms. studies on naturally occurring bacterial
Immunocompromised persons should be aware symbionts. Parasitol Res. 1995;81(2):98–102.
38 MedICal parasItology In the phIlIppInes

Naegleria spp.

Parasite Biology 10 to 35 µm but when rounded are usually 10


to 15 µm in diameter. In culture, trophozoites
N aegleria spp. are free-living protozoans
with two vegetative forms: an ameba
(trophozoite form), and a flagellate (swimming
may get over 40 µm. The cytoplasm is granular
and contains many vacuoles. The single nucleus
is large and has a large, dense karyosome and
form). A dormant cyst form is produced when
lacks peripheral chromatin.
conditions are not favorable. Transformation
Naegleria spp. are thermophilic organisms
from the trophozoite to the flagellate form
which thrive best in hot springs and other warm
may facilitate more rapid movement toward
aquatic environments. Both nonpathogenic and
food sources.
pathogenic forms exist. Only Naegleria fowleri
There are two forms of trophozoites of
has been reported to consistently cause disease
Naegleria fowleri: ameboid and ameboflagellate,
in humans, although some non-fowleri species
only the former of which is found in humans
may cause opportunistic infections.
(Plate 2.10). The ameboid trophozoites measure

Plate 2.10. Naegleria fowleri trophozoites in ameboid (left) and ameboflagellate (right) forms
(Accessed from www.dpd.cdc.gov/dpdx)

Naegleria fowleri has three stages, cysts, by penetrating the nasal mucosa and migrating
trophozoites, and flagellated forms, in its life to the brain via the olfactory nerves. N. fowleri
cycle. The trophozoites replicate by promitosis trophozoites are found in cerebrospinal fluid
(nuclear membrane remains intact) and can turn (CSF) and tissue, while flagellated forms are
into temporary non-feeding flagellated forms, occasionally found in CSF. Cysts are not seen
which usually revert back to the trophozoite in brain tissue (Figure 2.4).
stage. Trophozoites infect humans or animals
Chapter 2: protozoan Infections 39

Figure 2.4. Life cycle of Naegleria fowleri


(Accessed from www.dpd.cdc.gov/dpdx)
40 MedICal parasItology In the phIlIppInes

Pathogenesis and Clinical Manifestations shows a fibrinopurulent exudate consisting


mostly of neutrophils in the leptomeninges
N. fowleri is the causative agent
and brain tissue, and pockets of amebae with
of a rare but rapidly destructive and fatal
scant inflammatory exudates in necrotic areas.
meningoencephalitis termed primary amebic
Death usually occurs as a result of cerebral or
meningoencephalitis (PAM). In contrast to
cerebellar herniation as a result of increased
GAE which is predominantly an opportunistic
intracranial pressure.
infection, PAM usually occurs in previously
healthy adults with a history of swimming. Diagnosis
Therefore, in contrast to Acanthamoeba which
Diagnosis of PAM is usually suspected in
is largely an opportunistic organism, N. fowleri
persons with a compatible history of exposure
is considered a true pathogen.
and a rapidly progressive meningoencephalitis.
N. fowleri is able to survive in elevated
In the past, definitive diagnosis of PAM was
temperatures and reproduces rapidly in
based on demonstration of characteristic
temperatures above 30°C. Aside from naturally
trophozoites in the brain and cerebrospinal
occurring hot springs, warm geothermal plant
fluid. Aspirates from suspected infections, when
effluent into lakes and streams can lead to
introduced into bacteria-seeded agar culture
proliferation of amebae.
medium, will exhibit active trophozoites within
Most cases of PAM have occurred in young,
24 hours.
healthy persons who swim in contaminated
Naegleria trophozoites can be identified by
water. The route of entry is through invasion
the presence of blunt, lobose pseudopodia and
of organisms through the olfactory bulb after
directional motility. Flagellation tests have poor
accidental inhalation of water containing
sensitivity for identification since amebae which
the organisms. The sustentacular cells of the
test negative have been subsequently identified
olfactory neuroepithelium are thought to
as Naegleria spp. and Naegleria fowleri with more
phagocytose the amebae and transport these
sensitive and specific molecular techniques such
through the cribriform plate and into the brain.
as PCR and immunostaining. Serology utilizing
Multiple mechanisms then come into play,
ELISA is less useful in diagnosing active
producing a cytopathic effect on host tissues.
infection since healthy individuals especially in
These mechanisms include secretion of lytic
endemic areas have been shown to have positive
enzymes, membrane pore-forming proteins,
antibody titers.
factors which induce apoptosis, and direct
feeding on cells by the amebae. Treatment
In humans, PAM presents as fever, nausea,
Most persons infected with Naegleria die
vomiting, headache, nuchal rigidity, and mental
prior to institution of effective treatment.
status changes, with rapid progression to coma
Symptoms of PAM are indistinguishable from
and death. Characteristic cerebrospinal fluid
bacterial meningitis. Initial CSF results are
findings include elevated white blood cell count
suggestive of a bacterial etiology, and so patients
with neutrophilic predominance, high protein,
are typically treated with antibiotics which have
and low glucose.
no activity against Naegleria.
Post-mortem examination of infected brain
Amphotericin B in combination with
shows hemorrhagic necrosis, particularly of
clotrimazole is synergistic, and has been
the olfactory bulbs, congestion and edema of
successfully used to treat PAM. Amphotericin
neural tissue. Leptomeninges are inflamed and
B produces deleterious changes in the nucleus
congested as well. Microscopic examination
Chapter 2: protozoan Infections 41

and mitochondria of the ameba, decreases Prevention and Control


the number of food vacuoles, and increases
The ubiquitous nature of Naegleria, in
the formation of autophagic vacuoles. Ameba
contrast to the rarity of infection seems to
exposed to amphotericin B exhibit decreased
indicate that incidental exposure is unlikely to
pseudopod formation and form blebs on
lead to disease. Most instances of infection are
the plasma membrane. Newer agents such
related to invasion of the ameba through the
as azithromycin and voriconazole have been
olfactory bulbs, and so avoiding immersion
shown to be active against N. fowleri, both in
of the head and accidental inhalation of water
vitro and in vivo.
should be practiced in endemic areas and in hot
Epidemiology springs. No known cases of PAM have resulted
from drinking ameba-infected water.
Distribution of Naegleria in freshwater
Naegleria fowleri is easily killed by
lakes and ponds has been correlated with
chlorination of water at 1 ppm or higher.
physical, chemical, and biological parameters.
Infection has been reported from swimming
Strains have been frequently isolated from
in contaminated water with inadequate
thermal effluents, hot springs, and water with
chlorination, and so recommendations for
naturally or artificially elevated temperatures.
appropriate decontamination of swimming
Fecal coliform contamination provides a ready
water should be followed, especially in areas of
food source for ameba, and may increase the risk
high prevalence.
of infection due to higher density of organisms.
Studies on local Naegleria have identified References
a new species which is morphologically
Behets J, Seghi F, Declerck P, Verelst L, Duvivier
indistinguishable but biochemically distinct
L, Van Damme A, et al. Detection of
from other known species. Isolates from a
Naegleria spp. and Naegleria fowleri: a
thermally-polluted stream, an artificially-heated
comparison of flagellation tests, ELISA and
swimming pool, and from the brain aspirate of
PCR. Water Sci Technol. 2003;47(3):117–
a young patient have all yielded a single species,
22.
N. philippinensis. This has been extensively
Enriquez GL. Studies on Naegleria isolate
studied by Castro et al., and Matias et al. Only
from a reported case of PAM from the
one case of PAM has been reported locally, in
Philippines. 1989. Located at: College of
a young male with a history of swimming in
Public Health Library, University of the
fresh water. He responded well to amphotericin
Philippines Manila.
B infusion.
Matias RR, Enriquez GL, Schotellius J. Surface
Two Philippine isolates of Naegleria
lectin receptors on a Naegleria species from
(NSzu and RITM strains) have been evaluated
the Philippines. Lectins Biol Biochem Clin
for pathogenicity. Massive doses of amebae
Biochem. 1990;7:329–33.
successfully established infection in the brain
Visvesvara GS, Moura H, Schuster FL.
and caused death in some mice within two to
Pathogenic and opportunistic free-living
six days post-inoculation. Clinical features of
amoebae: Acanthamoeba spp., Balamuthia
infection and histopathology were compatible
mandrillaris, Naegleria fowleri, and Sappinia
with PAM.
diploidea. FEMS Immunol Med Microbiol.
2007;50:1–26.
42 MedICal parasItology In the phIlIppInes

Ciliates and Flagellates


Vicente Y. Belizario, Jr., Francis Isidore G. Totañes

Balantidium coli Human infection results from ingestion


of food and/or water contaminated with B.

I nitially identified as Paramecium coli by


Malmsten in 1857, Balantidium coli was
later described and placed under a separate
coli cysts. The incubation period is normally
from 4 to 5 days. Ingested cysts excyst in the
small intestines and become trophozoites.
genus in 1863. B. coli is the causative agent Trophozoites inhabit the lumen, mucosa, and
of the zoonotic disease called balantidiasis, submucosa of the large intestines, primarily
balantidiosis, or balantidial dysentery. It is the cecal region. They cause pathologic
considered as the largest protozoan parasite changes in the colonic wall and mucosa.
affecting humans and is the only ciliate Parasite reproduction occurs asexually through
known to cause human disease. It is capable of asymmetric binary fission, although sexual
attacking the intestinal epithelium, resulting in reproduction through conjugation has been
ulcer formation which, in turn, causes bloody reported. Cysts are formed principally as
diarrhea similar to that of amebic dysentery. protection for survival outside the host. The
This organism is primarily associated with pigs, parasites encyst during intestinal transport or
its normal host. after evacuation of semi-formed stools. Cysts
are the infective stage, and they may remain
Parasite Biology
viable for several weeks (Figure 2.5).
Balantidium coli trophozoite measures
Pathogenesis and Clinical Manifestations
30 to 150 µm long and 25 to 120 µm wide.
For locomotion, trophozoites are covered Balantidium coli trophozoites are capable of
with cilia arranged in a longitudinal pattern attacking the intestinal epithelium and creating
extending from the oral to the caudal region. a characteristic ulcer with a rounded base and
It has a cytostome, an oral apparatus at wide neck, in contrast to the flask-shaped,
the tapered anterior end, through which it narrow necked ulcers of amebiasis. Ulceration is
acquires food, and a cytopyge at the rounded caused by the lytic enzyme hyaluronidase which
posterior end through which it excretes waste. is secreted by the trophozoite. The trophozoites
It has two dissimilar nuclei. The macronucleus are abundant in exudates on mucosal surfaces;
is usually bean-shaped and can easily be while inflammatory cells and trophozoites are
identified in stained specimens, while the numerous in the base of the ulcers. Trophozoites
micronucleus is round and lies in the concavity also invade the submucosa and the muscular
of the macronucleus. B. coli has two contractile coat, including blood vessels and lymphatics.
vacuoles that act as osmoregulatory organelles. Intrinsic host factors including nutritional
The parasite also contains extrusive organelles status, intestinal bacteria flora, achlorhydria,
called mucocysts which are located beneath the alcoholism, and presence of chronic disease
cell membrane. contribute to host susceptibility to and severity
B. coli cysts are spherical to slightly ovoid of B. coli infection. It has been suggested by
in shape and measure 40 to 60 µm in diameter. some investigators that B. coli mucocysts might
They are covered with thick cell walls (double- have a function in the adhesion of parasitic
walled). Unlike amebae, encystation does not ciliates that may contribute to parasite virulence,
result in an increase in number of nuclei. although no definitive study has proven this. In
Chapter 2: protozoan Infections 43

Figure 2.5. Life cycle of Balantidium coli


(Accessed from www.dpd.cdc.gov/dpdx)
44 MedICal parasItology In the phIlIppInes

one study, it was shown that mucocysts in B. Diagnosis


coli trophozoites obtained from symptomatic
Diagnosis is made by microscopic
pigs were more numerous compared with
demonstration of trophozoites and cysts in
trophozoites obtained from asymptomatic hosts.
feces using direct examination or concentration
In addition, co-infection with other organisms
(sedimentation or flotation) techniques.
may also contribute to severity of B. coli infection.
Repeated stool examinations may be done to
The presence of Salmonella in the intestines
increase sensitivity. Demonstrating the presence
has been shown to aggravate balantidiasis by
of trophozoites in biopsy specimens from lesions
invading the ulcers caused by the protozoan.
obtained through sigmoidoscopy is likewise
Balantidiasis has three forms of clinical
diagnostic. Bronchoalveolar washings may
manifestations. Asymptomatic carriers are
also contain B. coli trophozoites in the case of
those who do not present with diarrhea or
pulmonary infection.
dysentery, but may serve as parasite reservoir
in the community. Fulminant balantidiasis, Treatment
or balantidial dysentery involves diarrhea with
The treatment of choice for balantidiasis
bloody and mucoid stools, which is sometimes
is tetracycline or metronidazole. Treatment in
indistinguishable from amebic dysentery. Acute
adults and older children is with tetracycline
cases may have 6 to 15 episodes of diarrhea per
500 mg or 40 mg/kg/dose divided in four doses
day accompanied by abdominal pain, nausea,
for 10 days. Tetracycline is contraindicated
and vomiting. This form of balantidiasis is
in children less than eight years of age and in
often associated with immunocompromised
pregnant women. Metronidazole 750 mg three
and malnourished states. The third form of
times daily, or 35 to 50 mg/kg body weight/
balantidiasis is the chronic form wherein
day in three divided doses, may be given for
diarrhea may alternate with constipation, and
5 days. Iodoquinol may also be given at 650
may be accompanied by nonspecific symptoms
mg, or 40 mg/kg/dose, divided in three doses
such as abdominal pain or cramping, anemia,
for 20 days. Other alternative treatments
and cachexia.
for balantidiasis include doxycycline and
B. coli can spread to extraintestinal sites
nitazoxanide. Currently there are no reports of
including the mesenteric nodes, appendix, liver,
B. coli exhibiting drug resistance.
genitourinary sites, pleura, and lungs. One
case report involved the detection of a cavitary Epidemiology
lesion in the right upper lobe of the lung on
chest radiograph in a patient who presented The distribution of B. coli is cosmopolitan
with hemoptysis. The patient had a history of and is more prevalent in areas with poor
insulin-dependent diabetes and organic farming sanitation, close contact with pigs or pig feces
using pig manure as fertilizer. Bronchoalveolar (e.g., farms, abattoirs), and in overcrowded
lavage revealed B. coli trophozoites. Another case institutions (e.g., asylum, orphanages, prisons).
presented with pulmonary hemorrhage and iron Warm and humid climates in tropical and
deficiency anemia, and revealed numerous B. subtropical countries can also contribute to
coli trophozoites by bronchial biopsy and lavage. the survival of cysts. High prevalence levels
Complications of balantidiasis include in pigs have been reported in regions in Latin
intestinal perforation and acute appendicitis. America and the Middle East, as well as in the
Cases of mortality related to balantidiasis Philippines, Papua New Guinea, and the West
were reported to be associated with intestinal Irian province of Indonesia.
hemorrhage and shock, intestinal perforation, There is an estimated 1% worldwide
or sepsis. prevalence of human B. coli infection. Pigs
Chapter 2: protozoan Infections 45

are the major host of balantidiasis, although Dodd LG. Balantidium coli infestation as a
primates have been reported to harbor infection. cause of acute appendicitis. J Infect Dis.
Prevalence studies in the United States and in 1991; 163:13–92.
Europe have reported infection rates ranging Goldberg JE, Parasitic colitides. Clin Colon
from 5% to as high as 100% in some areas. In Rectal Surg. 2007;20:38–46.
a study done in two (northern and southern) Karanis P, Kourenti C, Smith H. Waterborne
sites in the Philippines, an examination of pigs transmission of protozoan parasites: a
revealed 66.1% prevalence of B. coli infection. worldwide review of outbreaks and lessons
There has been a single report of an outbreak learnt. J Wat Health. 2007;5:1–38.
of balantidiasis that occurred in the Truk island Koopowitz A, Smith P, van Rensburg N,
in Micronesia in 1971. Rudman A. Balantidium coli-induced
pulmonary haemorrhage with iron
Prevention and Control
deficiency. S Afr Med J. 2010;100:534–6.
Control measures for balantidiasis include Ladas SD, Savva S, Frydas A, Kaloviduris
proper sanitation, safe water supply, good A, Hatzioannou J, Raptis S. Invasive
personal hygiene, and protection of food from balantidiasis presented as chronic colitis
contamination. Measures to limit contact of and lung involvement. Dig Dis Sci.
pigs with water sources and food crops may 1989;34(10):1621–3.
also contribute to reducing transmission La Via MV. Parasitic gastroenteritis. Pediatr
and infection. Use of pig feces as fertilizer Ann. 1994;23(70):556–60.
should also be avoided. Though cysts may Lee JL, Lanada EB, More SJ, Cotiw-an
be resistant to environmental conditions and BS, Taveros AA. A longitudinal study
may survive for long periods of time, they are of growing pigs raised by smallholder
easily inactivated by heat and by 1% sodium farmers in the Philippines. Prev Vet Med.
hypochlorite. Ordinary chlorination of water 2005;70:75-93.
is not effective against B. coli cysts. Nakauchi K. The prevalence of Balantidium coli
infection in fifty-six mammalian species. J
References
Vet Med Sci. 1999;61(1):63–5.
Beaver PC, Jung RC, Cupp EW. Clinical Nilles-Bije ML, Rivera WL. Ultrastructural and
parasitology. Philadelphia: Lea and Febiger; molecular characterization of Balantidium
1984. coli isolated in the Philippines. Parasitol
Belding DL. Textbook of parasitology. 3rd Res. 2010;106:387–94.
ed. New York: Appleton-Century Crofts; Sharma S, Harding G. Necrotizing lung infection
1965. caused by the protozoan Balantidium coli.
Borda CE, Rea MJ, Rosa JR, Maidana C. Can J Infect Dis. 2003;14(3):163–6.
Intestinal parasitism in San Cayetano, Schuster FL, Ramirez-Avila L. Current world
Corrientes, Argentina. Bull of Pan Am status of Balantidium coli. Clin Microbiol
Health Organ. 1996;30(3):227–33. Rev. 2008;21(4):626–38.
Farthing MJ. Treatment options for the Skotarczak B. Cytochemical identification of
eradication of intestinal protozoa. Nat mucocysts in Balantidium coli trophozoites.
Clin Pract Gastroenterol Hepatol. Folia Biol. 1999;47(1-2):61–5.
2006;3(8):436–45. The Medical Letter. Drugs for parasitic
infections [Internet]. Available from www.
medicalletter.org.
46 MedICal parasItology In the phIlIppInes

Giardia duodenalis
Juan Antonio A. Solon

G iardia duodenalis is an intestinal parasitic


flagellate of worldwide distribution. It is
known to cause epidemic and endemic diarrhea.
Cysts are ovoid and measure 8 to 12 µm
long by 7 to 10 µm wide. The young cysts have
two nuclei, while the mature cysts have four.
This protozoan is also known as Giardia Cysts are characterized by flagella retracted into
intestinalis or G. lamblia. It was first discovered axonemes, the median or parabasal body, and
in 1681 by Antoine van Leeuwenhoek in his deeply stained curved fibrils surrounded by a
own stools and was first described by Lambl in tough hyaline cyst wall secreted from condensed
1859 who called it Cercomonas intestinalis. It was cytoplasm.
later renamed Giardia lamblia by Stiles in 1915. Cysts from animals or human feces are
The disease caused by this parasite is called transferred to the mouth via contaminated
giardiasis, and this manifests as a significant hands, food, or water. Once mature cysts
but not life-threatening gastrointestinal disease. (infective stage) are ingested, they pass safely
through the stomach and excyst in the
Parasite Biology
duodenum (in about 30 minutes) developing
Giardia duodenalis is a flagellate that lives into trophozoites which rapidly multiply and
in the duodenum, jejunum, and upper ileum attach to the intestinal villi causing pathologic
of humans. It has a simple asexual life cycle changes. The trophozoites may then be found
that includes trophozoites and quadrinucleated in the jejunum. As the feces enters the colon
infective cyst stages. Molecular typing of isolates and dehydrates, the parasite then encysts. After
shows that those which parasitize humans can encystment, mature cysts are passed out in the
be classified as belonging to either assemblage feces and are infectious (Figure 2.6).
A or B genotypes based on specific sequences
Pathogenesis and Clinical Manifestations
in the small subunit of their ribosomal RNA.
The trophozoites measure 9 to 12 µm Infection with G. duodenalis occurs when
long by 5 to l5 µm wide. They are pyriform the host ingests food or water contaminated
or teardrop shaped, pointed posteriorly, with with the mature cysts. Depending on the strain
a pair of ovoidal nuclei, one on each side of involved, infection can occur with one ingesting
the midline. The dorsal side of the organism is as few as 10 cysts. The ability of the parasite to
convex, while the ventral side is concave with cause disease can be traced to its ability to alter
a large adhesive disc used for attachment. It is mucosal intestinal cells once it has attached to
bilaterally symmetrical, with a distinct medial the apical portion of the enterocyte. The parasite
line called the axostyle. The parasite is propelled attaches to the intestinal cells via an adhesive
into an erratic tumbling motion by four pairs sucking disc located on its ventral side, causing
of flagella arising from superficial organelles in mechanical irritation in the affected tissues.
the ventral side of the body. Trophozoites divide Several studies have investigated this mechanism
by longitudinal binary fission and are found in of attachment. In monolayer studies, it was
diarrheic stools. Antigenic variation results in noted that attachment was influenced by certain
the entire surface of the parasite being covered physical factors such as temperature and pH.
with variant-specific surface proteins (VSPs). Attachment was observed to be maximal at
Chapter 2: protozoan Infections 47

Figure 2.6. Life cycle of Giardia duodenalis


(Accessed from www.dpd.cdc.gov/dpdx)
48 MedICal parasItology In the phIlIppInes

body temperature and stable at a pH of 7.8 be asymptomatic. For acute cases, patients
to 8.2. The parasite may also produce a lectin experience abdominal pain, described as
which, when activated by duodenal secretions, cramping, associated with diarrhea. There is
is able to facilitate attachment. Once attached, also excessive flatus with an odor of “rotten
the organism is able to avoid peristalsis by eggs” due to hydrogen sulfide. Other clinical
trapping itself in between the villi or within the features include abdominal bloating, nausea,
intestinal mucus. and anorexia. Diarrhea is the most common
Upon attachment to the intestinal cells, symptom, occurring in 89% of cases. It is
G. duodenalis is able to cause alterations in followed by malaise and flatulence. Spontaneous
the villi such as villous flattening and crypt recovery occurs within 6 weeks in mild to
hypertrophy. These alterations lead to decreased moderate cases. In untreated cases, patients may
electrolyte, glucose, and fluid absorption, and experience diarrhea with varying intensities, for
cause deficiencies in disaccharidases. Studies on weeks or months.
Giardia muris-infected mice showed diffuse loss Chronic infection is characterized by
of microvillous surface area which investigators steatorrhea, or the passage of greasy, frothy
also correlated to decreased maltase and stools. In some cases, periods of diarrhea have
sucrase activities. The physiologic disturbances been observed to alternate with normal or even
subsequently result in malabsorption and constipated bowel periods. There may be weight
maldigestion, which in turn cause the signs and loss, profound malaise, and low-grade fever. In
symptoms experienced by the patient. Bacterial developing countries, it has been described as a
colonization of the area may further worsen the cause of the failure-to-thrive syndrome.
damage already caused by the parasite.
Diagnosis
In other studies, G. duodenalis was shown
to rearrange the cytoskeleton in human colonic Diagnosis is made by demonstration of G.
and duodenal monolayers. Cytoskeleton is duodenalis trophozoites (Plate 2.11) and/or cysts
essential for proper cell attachment to the (Plate 2.12) in stool specimens. Trophozoites
extracellular matrix and the other neighboring in direct fecal smears may be characterized as
cells. Changes observed in apoptotic cells having a floating leaf-like motility. To detect
include disruption of the cytoskeleton that leads
to structural disintegration and detachment
from the substrate. Hence, the parasite has
been suggested to cause enterocyte apoptosis.
This finding was strengthened by another study,
which showed the ability of the parasite not
only to disrupt cellular tight junctions but also
to increase epithelial permeability, thus, leading
to the loss of epithelial barrier function. With
this loss of barrier function, luminal contents
may penetrate the submucosal layers causing
more damage in the intestinal tissue.
From ingestion of the cysts, it takes about Plate 2.11. Giardia duodenalis trophozoite
1 to 4 weeks (average of 9 days) for the disease (Courtesy of the Department of Parasitology,
to manifest. Half of the infected patients may UP-CPH)
Chapter 2: protozoan Infections 49

the highest combination of sensitivity and


specificity.
Treatment

Giardiasis may be treated with


metronidazole 250 mg three times a day for 5
to 7 days (pediatric dose: 15 mg/kg/day in three
divided doses). Metronidazole is usually well-
tolerated in adults and has a cure rate of 90%.
Alternative drugs include tinidazole (single
dose of 2 g for adults; 50 mg/kg in children)
and furazolidone (100 mg four times daily
Plate 2.12. Giardia duodenalis cysts for 10 days for adults; 6 mg/kg/day in four
(Courtesy of the Department of Parasitology, divided doses for 7 to 10 days). Albendazole
UP-CPH) is an alternative at 400 mg/day for 5 days in
adults and 10 mg/kg/day for 5 days in children.
cysts in stools, concentration techniques are A meta-analysis has shown that albendazole is
recommended. At least three stool examinations equally effective as metronidazole at the above
on alternate days are recommended because of doses. Although not available in the Philippines,
spotty shedding of cysts. If the parasite is not nitazoxanide has likewise been used effectively
found in the feces, duodeno-jejunal aspiration in drug-resistant cases.
may be done. Examination of the duodenal Prompt treatment of asymptomatic
contents for trophozoites gives a higher individuals reduce cyst passage and possible
percentage of positive findings compared to transmission especially among high risk groups
examination of feces. In a patient with chronic such as food handlers, institutionalized patients,
diarrhea, giardiasis should be considered as a children attending day-care, and day-care
possible cause. workers.
Aside from duodenal aspiration, the
Epidemiology
Enterotest® (HDC Mountain View, CA) may
demonstrate Giardia trophozoites. The patient Giardia has a worldwide distribution. In
swallows a gelatin capsule attached to a nylon the Philippines, the prevalence of giardiasis
string, with one end of the string attached to ranges from 1.6 to 22.0% depending on the
the patient’s cheek. After about 4 to 6 hours, population group being studied. From the local
the string is removed, and any adherent fluid is data, it can be clearly seen that the groups in
placed on the slide for microscopic examination. areas with poor sanitation and hygiene practices
Presently, antigen detection tests and have a higher prevalence of giardiasis.
immunofluorescent tests are already available Notably, Giardia is not commonly found
as commercial kits. Immunochromatographic in patients with diarrhea. In this symptomatic
assays detect the presence of Giardia antigen population, the prevalence was similar between
in stool. Cyst wall protein 1 (CWP1) is one children (<18 years old) and adults. However,
of the antigens used for these diagnostic tests. the prevalence of giardiasis was significantly
Direct fluorescent antibody assays have been higher in male adults than females, a trend also
considered by many laboratories as the gold seen in other countries (Table 2.2).
standard in diagnosis as such assays have
50 MedICal parasItology In the phIlIppInes

Table 2.2. Selected Philippine data on giardiasis

Reference Population (n) Prevalence


Cross and Basaca-Sevilla (1986) Community (n=30,000) 6.0%
Auer (1990) Urban poor, 8 months – 15 years (n=238) 20.0%
Bustos, et al. (1991) Mentally ill patients (n=176) 17.0%
Lee, et al. (1999) Children (Legazpi City) n=64 7.8%
Belizario, et al. (2000) Patients from the community suspected with capillariasis
4.2%
(Brgy. Awao, Compostela Valley) (n=72)
Belizario, et al. (2000) Community (Brgy. San Isidro, Compostela Valley) (n=242) 7.4%
Avila, et al. (2003) Food handlers, school canteen (n=123) 3.3%
Esparar, et al. (2003) Food handlers, tertiary hospital (n=59) 3.4%
Kim, et al. (2003) Community (n=301) 0.0%
Baldo, et al. (2004) Institutionalized children (Metro Manila) (n=172) 11.6%
Belizario, et al. (2005) Mall employees (Cebu city) (n=256) 0.8%
Natividad, et al. (2008) Diarrheic patients (n=3,456) 2.0%
UP-CPH Department of Parasitology Referred patients (n=667)
<1.0%
Laboratory (2006-2010)
Yason and Rivera (2007) Urban poor (n=2,354) 22.0%

The first published study on Giardia or by infected food handlers. Normal water
genotypes in the Philippines showed that the chlorination will not affect cysts, but usual
majority (86%) of the isolated genotypes belong water treatment modalities should be adequate.
to assemblage B.
References
Direct oral-anal sexual contact among men
who have sex with men may increase the risk Adam RD. Biology of Giardia lamblia. Clin
of giardiasis and infection with other intestinal Microbiol Rev. 2001;14(3):447–75.
protozoans. Adam RD, Nigam A, Seshadri V, Martens
Outbreaks of giardiasis are more frequently CA, Farneth GA, Morrison HG, et al.
reported outside the Philippines. Most of these The Giardia lamblia vsp gene repertoire:
are water-borne (recreational water or drinking characteristics, genomic organization, and
water). Foodborne outbreaks have also been evolution. BMC Genomics. 2010;11:424.
reported. The low infective dose, prolonged Auer C. Health status of children living in
communicability, and relative resistance to a squatter area of Manila, Philippines,
chlorine facilitate the transmission of Giardia with particular emphasis on intestinal
through drinking and recreational water, food, parasitoses. Southeast Asian J Trop Med
and person-to-person contact. Pub Health. 1990;21(2):289–300.
Prevention and Control
Avila MS, Garcia MR, Narcelles MV, Serra
FB, Tejida BM. Prevalence of intestinal
Methods of prevention and control include helminth and protozoan infections among
proper or sanitary disposal of human excreta food-handlers in selected school canteens
to prevent contamination of food and water in Manila [undergraduate special study].
supply. The former can be contaminated 2003. Located at: College of Public Health,
by the use of night soil as fertilizer, by flies, University of the Philippines Manila.
Chapter 2: protozoan Infections 51

Baldo ET, Belizario VY, de Leon WU, Kong Esparar DG, Belizario VY, Jr., Relos JR.
HH, Chung DI. Infection status of Prevalence of intestinal parasitic infections
intestinal parasites in children living in among food handlers of a tertiary hospital
residential institutions in Metro Manila, in Manila using direct fecal smear and
the Philippines. Korean J Parasitol. formalin ether concentration technique.
2004;42(2):67–70. Phil J Microbiol Infect Dis. 2004;33(3):1–
Beaver PC, Jung RC, Cupp EW. Clinical 6.
parasitology. 3rd ed. Philadelphia: Lea and Faubert G. Immune Response to Giardia
Febiger; 1984. d u o d e n a l i s . C l i n Mi c r o b i o l Re v.
Belding DL. Textbook of parasitology. New 2000;13(1):35–54.
York: Appleton-Century Crofts; 1965. Gardner TB, Hill DR. Treatment of Giardiasis.
Belizario VY, Jr., de Leon WU, Esparar Clin Microbiol Rev. 2001;14(1):114–28.
DG, Galang JM, Fantone J, Verdadero Jones JE. String test for diagnosing Giardiasis.
C. Compostela Valley: a new endemic Am Fam Physician. 1986;34(2):123–6.
focus for Capillariasis philippinensis. Kim BJ, Ock MS, Chung DI, Yong TS, Lee
Southeast Asian J Trop Med Pub Health. KJ. The intestinal parasite infection
2000;31(3):478–81. status of inhabitants in the Roxas City,
Belizario VY, Jr., Bersabe MJ, de Leon WU, the Philippines. Korean J Parasitol.
Bugayong MG, de Guzman AD, et al. A 2003;41:113–5.
new look at heterophyidiasis (intestinal Korman SH, Hais E, Spira DT. Routine in
fluke infection): a food-borne parasitic vitro cultivation of Giardia lamblia by
zoonosis in the Philippines. In: Department using the string test. J Clin Microbiol.
of Health research compendium 1993– 1990;28(2):368–9.
2001. Manila (Philippines): Department Kuntz RE. Intestinal parasites of man in the
of Health; 2001. p. 60–1. republic of the Philippines. J Philipp Med
Belizario VY, Diaz AB, Esparar DG, Bugayong Assoc. 1963;39(7):590–600.
MG. Parasitologic screening of mall Lee KJ, Ahn YK, Yong TS. A small-scale
employees applying for health certificates survey of intestinal parasite infections
at a local health office in Cebu City: among children and adolescents in Legaspi
implications for policy and practice. Phil J City, the Philippines. Korean J Parasitol.
Microbiol Infect Dis. 2005;34(2):65–70. 2000;38:183–5.
Bustos MD, Salazar N, Espino FE, Montalban Nash TE. Antigenic variation in Giardia
CS, Sabordo D, Laurente M. Ornidazole in lamblia and the host’s immune response.
the treatment of giardiasis in an institution Philos Trans R Soc Lond B Biol Sci.
for the mentally retarded. Phil J Microbiol 1997;352(1359):1369–75.
Infect Dis. 1991;20(1):13–6. Nash TE, Herrington DA, Losonsky GA,
Cross JH, Sevilla VB. Biomedical surveys in Levine MM. Experimental human
the Philippines. Manila (Philippines): US infections with Giardia lamblia. J Infect
NAMRU Unit No. 2; 1984. Dis. 1987;156(6):974–84.
Dib HH, Lu SQ, Wen SF. Prevalence of Giardia Natividad FF, Buerano CC, Lago CB,
lamblia with or without diarrhea in South Mapua CA, de Guzman BB, Seraspe
East, South East Asia and the Far East. EB, et al. Prevalence rates of Giardia and
Parasitol Res. 2008;103(2):239–51. Cryptosporidium among diarrheic patients
52 MedICal parasItology In the phIlIppInes

in the Philippines. Southeast Asian J Trop Yason JA, Rivera WL. Genotyping of Giardia
Med Public Health. 2008;39(6):991–9. duodenalis isolates among residents of slum
Oberhuber G, Stolte M. Giardiasis: analysis of area in Manila, Philippines. Parasitol Res.
histological changes in biopsy specimens of 2007;101(3):681–7.
80 patients. J Clin Pathol. 1990;43(8):641– Yoder JS, Harral C, Beach MJ. Giardiasis
3. surveillance—United States, 2006-2008.
Upcroft P, Upcroft JA. Drug targets and MMWR Surveill Summ. 2010;59(6):15–
m e c h a n i s m s o f re s i s t a n c e i n t h e 25.
anaerobic protozoa. Clin Microbiol Rev.
2001;14(1):150–64.
Chapter 2: protozoan Infections 53

Trichomonas vaginalis
Juan Antonio A. Solon

T richomonas vaginalis causes a sexually


transmitted disease called trichomoniasis
which has a worldwide distribution. Its
of the tissue layer. The trophozoites infect the
surface but do not appear to invade the mucosa.
The acute inflammation caused by the parasite
incidence correlates strongly with the number results in the characteristic liquid vaginal
of sexual partners. It was first observed by secretions, greenish or yellow in color, that
Donne in 1836 in purulent secretions of male cover the mucosa down to the urethral orifice,
and female urogenital tracts. It is now often vestibular glands, and clitoris. The vaginal
described as the most prevalent non-viral secretions are very irritating and may cause
sexually transmitted infection. intense itchiness and burning sensation. As the
acute condition changes to the chronic stage, the
Parasite Biology
secretion loses its purulent appearance due to a
Trichomonas vaginalis exists only in the decrease in the trichomonads and leukocytes, an
trophozoite stage. It has a pyriform shape, increase in epithelial cells, and the establishment
measuring 7 to 23 µm with four free anterior of a mixed bacterial flora. Aside from the
flagella that appear to arise from a simple common symptoms of vaginal discharge,
stalk, and a fifth flagellum embedded in the vulvitis, and dysuria, trichomonads appear to
undulating membrane. This membrane extends be associated with an increased incidence of
to about half the organism’s length. The parasite postpartum endometritis. Complications in
has a median axostyle and a single nucleus. women include secondary bacterial infection
The parasite is found in the urogenital of the urogenital tract.
tract. In women, it is found in the vagina but Speculum examination reveals punctate
may ascend as far as the renal pelvis. The parasite hemorrhages of the cervix, the so-called
can be isolated from the urethra, prostate, and strawberry cervix, which is observed in only
less frequently, in the epididymis in men. The 2% of cases.
trophozoites multiply by binary fission in the Trichomonas infection in males may be
host and are transferred passively from person latent and essentially asymptomatic. In some
to person (Figure 2.7). The usual mode of cases, it is responsible for an irritating persistent
transmission is by sexual intercourse. and recurring urethritis. Prostatitis is the most
common complication.
Pathogenesis and Clinical Manifestations
Diagnosis
Inflammation of the vaginal mucosa occurs
several days after the inoculation of T. vaginalis Saline preparation of vaginal fluid is
trophozoites. T. vaginalis cannot live without the quickest and most inexpensive way to
close association with the vaginal, urethral, diagnose trichomoniasis, but the sensitivity
or prostatic tissues. Four to 28 days after of this technique is low at 60 to 70%. The
introduction of viable T. vaginalis into the vagina, accepted gold standard is culture which
proliferating colonies of the flagellate cause takes 2 to 5 days. The unstained wet drop
degeneration and desquamation of the vaginal preparations may be fixed and stained by
epithelium followed by leukocytic inflammation Giemsa, Papanicolau, Romanowsky, and
54 MedICal parasItology In the phIlIppInes

Figure 2.7. Life cycle of Trichomonas vaginalis


(Accessed from www.dpd.cdc.gov/dpdx)

acridine orange stains. Trichomonas can also be detection tests and polymerase chain reaction
cultured using Diamond’s modified medium, (PCR) assays are commercially available, but
and Feinberg and Whittington culture medium. not widely used locally. PCR among females
The Pap smear may also show trichomonads does not seem to offer an added diagnostic
(sensitivity 60%; specificity 95%). Antigen advantage. Among males, however, diagnosis is
Chapter 2: protozoan Infections 55

more difficult. For culture, the best results are Epidemiology


seen with a combination of cultures of urethral
Trichomonas infection occurs worldwide. It
swabs and urine sediment. PCR appears to
is estimated that there are 170 to 190 million
detect more cases than culture among males.
individuals with trichomoniasis. Prevalence is
The InPouchTM TV Test is a novel transport
higher among women of child-bearing age.
and culture test system which allows the
About 5 to 20% of women and 2 to 12% of
specimen to be inoculated into a sealed pouch
men in developed countries are infected. Higher
with culture media. Growth can be monitored
prevalence is associated with greater frequency
microscopically directly through the pouch.
of sexual intercourse with multiple partners and
This test has a comparable sensitivity to
with commercial sex workers. Trichomoniasis is
Diamond’s modified medium culture.
often associated with other sexually transmitted
Treatment infections. In a study in the United States, 70%
of male partners of women with trichomoniasis
Trichomoniasis can be treated with
were likewise infected and the majority of the
metronidazole or tinidazole 2 g as a single
infected male partners were asymptomatic
dose. The reported cure rates of these drugs
(77%).
range from 86 to 100%. Sexual partners must
In the Philippines, the prevalence of
be treated concomitantly to prevent reinfection.
trichomoniasis among commercial sex workers
If metronidazole treatment failure occurs and
varies with the method of diagnosis used,
reinfection is ruled out, a seven-day regimen of
from 15% in studies using only microscopic
500 mg metronidazole three times a day may
examination of vaginal swabs to 37% in studies
be considered. If either this regimen fails, a 2
using culture. One study surveyed 421 male
g daily dose for 5 days of either metronidazole
sex workers and there were no positive cases
or tinidazole can be used. In pregnancy,
among them based on microscopy (Table
metronidazole remains the drug of choice for
2.3). Local isolates of T. vaginalis have been
trichomoniasis.
Table 2.3. Selected Philippine studies on trichomoniasis

Reference Population (n) Method Prevalence


Arambulo, et al. (1977) Waitresses/hostesses and Microscopy Among waitresses/hostesses: 15.0%
housewives (n=560) Housewives: 2.7%
Overall: 5.9%
Basaca-Sevilla, et al. (1986) Total=1371 Microscopy and 24.0% on initial examination; 37.0%
commercial sex workers culture after 5 days of culture
(n=1,284)
expectant mothers (n =87)
Jueco, et al. (1998) n=368 (women) Microscopy Overall prevalence: 12.0%

150 women from a private 8.0% from private clinic


gynecologic clinic
(housewives, workers,
students, vendors, factory
workers, business women,
beautician)

218 commercial sex workers Among sex workers: 14.8%


from a social hygiene clinic
Monzon, et al. (1991) Total=1,357 commercial sex Microscopy Females: 3.8%
workers; Males: 0.0%
females (n=936)
males (n=421)
56 MedICal parasItology In the phIlIppInes

characterized molecularly showing low genetic Gumbo FZ, Duri K, Kandawasvika GQ,
polymorphism. Kurewa NE, Mapingure MP, Munjoma
It is relevant to discuss trichomoniasis in the MW, et al. Risk factors of HIV vertical
context of HIV. In Zimbabwe and South Africa, transmission in a cohort of women
trial participants diagnosed with trichomoniasis under a PMTCT program at three peri-
were more likely to test positive for HIV in their urban clinics in a resource-poor setting. J
next visit. Perinatal transmission of HIV was Perinatol. 2010;30(11):717–23.
likewise more likely if the mother had vaginal Ju e c o N L , A r a n e t a C A , Ta d i n a E G .
infections. Epidemiology of Trichomonas vaginitis
among selected group of women in Manila.
Prevention and Control
Acta Med Philipp. 1988;24(3):85–6.
Prevention is best achieved by reducing Mavedzenge SN, Pol BV, Cheng H, Montgomery
the risk of exposure. Limiting the number of ET, Blanchard K, de Bruyn G, et al.
sexual partners, and proper use of protective Epidemiological synergy of Trichomonas
devices such as condoms and spermicidal foams vaginalis and HIV in Zimbabwean and
may help prevent infection. To prevent “ping- South African women. Sex Transm Dis.
pong” or recurrent infections, there should 2010;37(7):460–6.
be simultaneous treatment of sexual partners. Monzon OT, Santana RT, Paladin FJ, Bautista
Prompt follow-up of patients and their contacts, A, Fajutagana L, Eugenio S. The Prevalence
as well as health and sex education about of sexually transmitted diseases (STDs) and
venereal disease are also important. human immunodeficiency virus (HIV)
infection among Filipino sex workers. Phil
References
J Microbiol Infect Dis. 1991;20:41–4.
Arambulo PV, Cabrera BD, Osteria TS, Baltazar Rivera WL, Ong VA, Masalunga MC. Molecular
JC. A comparative study of Trichomonas characterization of Trichomonas vaginalis
vaginalis prevalence in Filipino women. isolates from the Philippines. Parasitol Res.
Southeast Asian J Trop Med Public Health. 2009;106(1):105–10.
1977;8:298. Schwebke JR, Burgess D. Trichomoniasis. Clin
Basaca-Sevilla V, Cross JH, Alquiza L, Lacap T. Microbiol Rev. 2004;17(4):794–803.
Prevalence of Trichomonas vaginalis in some Spence MR, Harwell TS, Davies MC, Smith JL.
Filipino women. Southeast Asian J Trop The minimum single oral metronidazole
Med Public Health. 1986;17(2):194–6. for treating trichomoniasis: a randomized,
Beaver PC, Jung RC, Cupp EW. Clinical blinded study. Obstet Gynecol. 1997;89(5):
parasitology. 9th ed. Philadelphia: Lea and 699–703.
Febiger; 1984. Van der Pol B. Trichomonas vaginalis infection:
Belding DL. Textbook of parasitology. New the most prevalent nonviral sexually
York: Appleton-Century Crofts; 1965. transmitted infection receives the least
Cudmore SL, Delgaty KL, Hayward-McClelland public health attention. Clin Infect Dis.
SF, Petrin DP, Garber GE. Treatment 2007;44(1):23–5.
of infections caused by metronidazole- Wendel KA, Workowski KA. Trichomoniasis:
resistant Trichomonas vaginalis. Clin challenges to appropriate management.
Microbiol Rev. 2004;17(4):783–93. Clin Infect Dis. 2007;44(Suppl 3):123–9.
Gerbase AC, Rowley JT, Mertens TE. Global
epidemiology of sexually transmitted
diseases. Lancet. 1998;351(Suppl 3):2–4.
Chapter 2: protozoan Infections 57

Non-Pathogenic Flagellates
Juan Antonio A. Solon

Trichomonas hominis Diagnosis is made by swabbing the tartar


between the teeth, the gingival margin, or

A s with other Trichomonas species, T. hominis


occurs only as a trophozoite which has a
pyriform shape and measures 7 to 13 µm. It has
tonsillar crypts.
Pulmonary trichomoniasis has been
reported among those with underlying chronic
five anterior flagella and a posterior flagellum pulmonary disease, entering the lungs most
projecting from an undulating membrane. The probably by aspiration. The parasite is probably
cytostome and the nucleus are situated at the unable to cause disease on its own. The presence
anterior end. An axostyle extends from anterior of bacteria most probably allows it to proliferate
to posterior along the mid-axis. Transmission profusely. In most of these cases, treatment with
occurs rapidly through fecal contamination of metronidazole results in rapid improvement.
food and drinks.
Its habitat is the cecal area of the large Chilomastix mesnili
intestine of human and other primates. It is non-
invasive. Trophozoites pass out with diarrheic This organism inhabits the cecal region
stools. The prevalence in the Philippines is less of the large intestine. It has well-defined
than 1%. trophic and cystic stages. The trophozoite is
asymmetrically pear-shaped as a result of a
Trichomonas tenax spiral groove extending through the middle half
of the body. Its size ranges from 6 to 10 µm.
Trichomonas tenax is a pyriform flagellate The characteristic boring and spiral forward
which has been observed only in the trophozoite movement is made possible by the three anterior
stage. It measures 5 to 12 µm, and is smaller and free flagella and a more delicate one within the
more slender than T. vaginalis. It has four free prominent cytostome.
equal flagella and a fifth one on the margin of The cyst is pear- or lemon-shaped, broadly-
an undulating membrane which does not reach rounded at one end and somewhat bluntly-
the posterior end of the body, and lacks a free conical at the other end which has a knob-like
posterior extension. It has a single nucleus and protruberance that is visible occasionally.
a cytostome. The organism multiplies by binary Internally, hematoxylin and eosin stained films
fission and thrives on the microorganisms found clearly demonstrate the single large vestibular
in its environment. nucleus and the cytostome, which is almost
Exposure results from droplet spray as long as the encysted organism. Good
from the mouth, kissing, or common use of preparations reveal a fibril on either side of the
contaminated dishes and drinking glasses. cytostome.
Trichomonas tenax is a harmless commensal of Transmission occurs through ingestion
the human mouth, living in the tartar around of cysts in food and drinks. Prevalence in
the teeth, in cavities of carious teeth, and in the Philippines is less than 1%. This is a
necrotic mucosal cells in the gingival margins. It harmless commensal diagnosed by microscopic
is quite resistant to changes in temperature and examination of feces and demonstration of
will survive for several hours in drinking water. either trophozoites or cysts. No treatment is
58 MedICal parasItology In the phIlIppInes

indicated. Preventive and control measures Southeast Asian J Trop Med Public Health.
include improved sanitation and personal 1981;12(1):12–8.
hygiene. Carney WP, de Veyra VU, Cala EM, Cross
JH. Intestinal parasites of man in
References
Bukidnon, Philippines, with emphasis on
Beaver PC, Jung RC, Cupp EW. Clinical schistosomiasis. Southeast Asian J Trop
parasitology. 9th ed. Philadelpha: Lea and Med Public Health. 1981;12(1):24–9.
Febiger; 1984. Cross JH, Banzon T, Wheeling CH, Cometa
Carney WP, Banzon T, de Veyra VU, Dana E, H, Lien JC, Clarke R, et al. Biomedical
Cross JH. Intestinal parasites of man in survey in North Samar Province, Philippine
Northern Bohol, Philippines, with emphasis Islands. Southeast Asian J Trop Med Public
on schistosomiasis. Southeast Asian J Trop Health. 1977;8(4):464–75.
Med Public Health. 1980;11(4):473–9. Hersh SM. Pulmonary trichomoniasis and
Carney WP, Banzon T, de Veyra VU, Papasin Trichomonas tenax. J Med Microbiol.
MC, Cross JH. Intestinal parasites of 1985;20(1):1–10.
man in Oriental Mindoro, Philippines, Lewis KL, Doherty DE, Ribes J, Seabolt JP,
with emphasis on schistosomiasis. Bensadoun ES. Empyema caused by
Trichomonas. Chest. 2003;123(1):291–2.
Chapter 2: protozoan Infections 59

Coccidians
Winifreda U. de Leon

T he coccidian parasites are the largest group


of apicomplexan protozoa falling under
Class Conoidasida. Coccidia is a subclass of
reported that the only species that infect
mammals was C. parvum and was believed
to be the species infecting humans. However,
microscopic, spore-forming, single-celled molecular tools, especially DNA analysis,
obligate intracellular protozoan. Members described the existence of another species,
of Phylum Apicomplexa are provided with Cryptosporidium hominis found mainly in
a cluster of secretory organelles made up of humans.
rhoptries, micronemes, and polar rings with
Parasite Biology
microtubules. In some species, a conoid may
be found within the polar rings as well. The All stages of development are completed in
secretion allows the parasite to enter the host the gastrointestinal tract of the host. Oocysts
cell. when passed out are already infective. Oocysts
Coccidians infect the intestinal tract of produced by C. hominis are found in the feces
most phyla of invertebrates and all classes of of humans and other animals. The oocysts are
vertebrates including humans. They fall under round and measure 4 to 5 µm in diameter.
Order Eucoccidiorida Suborder Eimeriorina. Each oocyst contains four sporozoites, which
The disease called coccidiosis is recognized as are present at the time of passage into the feces.
one of the major problems in animal farming The oocyst is infectious and when ingested, the
and in zoo management. Among humans, sporozoites attach to the surface of epithelial
they are considered to be opportunistic in cells of the gastrointestinal tract. The sporozoites
immunocompromised and immunodeficient develop into small trophozoites and become
individuals. Species with medical and veterinary intracellular but extracytoplasmic, and attach
significance include Cryptosporidium, Cyclospora, to the brush borders. The trophozoites divide
Cystoisospora, Sarcocystis, and Toxoplasma. by schizogony producing merozoites that infect
In the coccidian life cycle, there is an other cells. Macro- and microgametocytes are
alternation of sexual and asexual multiplication. eventually produced, and the macrogamete
It is typically characterized by three sequential is fertilized by the microgamete to produce
stages, namely: sexual cycle or sporogony a zygote. There are two types of oocysts
producing oocysts, asexual cycle or schizogony resulting from the zygote: the thin-walled
(merogony) producing merozoites (meronts), and the thick-walled oocysts. The thin-walled
and gametogony resulting in the development oocysts infect other enterocytes thus resulting
of male (micro) and female (macro) gametocytes in autoinfection, which is possibly responsible
(gamonts). The complexity in the life cycles of for the chronicity of the infection among the
coccidians is a challenge in terms of taxonomy. immunocompromised. On the other hand, the
thick-walled oocysts are passed out with the
Cryptosporidium hominis feces that may contaminate food and water,
which are ingested by the same or another host
There are several species of Cryptosporidium
(Figure 2.8).
that are currently recognized. It was initially
60 MedICal parasItology In the phIlIppInes

Figure 2.8. Life cycle of Cryptosporidium spp.


(Accessed from www.dpd.cdc.gov/dpdx)
Chapter 2: protozoan Infections 61

Pathogenesis and Clinical Manifestations Treatment

Cryptosporidiosis hominis was not well There is presently no acceptable treatment


recognized prior to the occurrence of acquired for cryptosporidiosis. Nitazoxanide, however,
immune deficiency syndrome (AIDS). In has been reported effective in preliminary trials.
the immunocompetent host, the disease may Bovine colostrum as well as paromomycin and
present as a self-limiting diarrhea lasting for clarithromycin have shown promise in treating
2 to 3 weeks, and less commonly, abdominal severe diarrhea. Azithromycin may also be of
pain, anorexia, fever, nausea, and weight loss. value. In addition to chemotherapy, body fluid
In immunocompromised persons, the diarrhea replacement and symptomatic treatment are
becomes more severe, progressive, and may recommended for both the immunocompetent
become life-threatening. The bile duct and gall and immunosuppressed patients.
bladder may become heavily infected and lead to
Epidemiology
acute and gangrenous cholecystitis. Respiratory
infections lead to chronic coughing, dyspnea, Cryptosporidiosis hominis has a universal
bronchiolitis, and pneumonia. distribution with infections reported worldwide.
The villi of the intestines become blunted Epidemics are unusual in North America,
and there is infiltration of inflammatory cells although there was a report of an epidemic
into the lamina propria and elongated crypts. involving over 400,000 cases in the state
There may be varying degrees of malabsorption of Wisconsin in the United States. This
and excessive fluid loss in immunocompromised epidemic was attributed to the use of a faulty
patients. Death may occur in disseminated water purification system. Most epidemics
infections. are associated with water, and in many cases,
the water was contaminated with calf feces.
Diagnosis
Cryptosporidium parvum of calves has been
There are several methods of stool reported to cause infection among veterinary
examination that will reveal C. hominis oocyst. attendants and visitors in dairy farms and
Sheather’s sugar flotation and the formalin petting zoos.
ether/ethyl acetate concentration technique Swimming in contaminated recreation
are commonly used. Kinyoun’s modified acid- water may result in accidental ingestion of
fast stain is routinely used with the oocysts infective oocysts. Swimming pool disinfection
appearing as red-pink doughnut-shaped circular with 3 to 5 mg/L of chlorine does not kill
organisms in a blue background. Intestinal the oocysts. The most common mode of
biopsy material may also be examined under a transmission is from one person to another.
light microscope and stages of the parasite can Infected food handlers may likewise transmit
be seen at the microvillus region of the infected oocysts during handling of beverages, raw
enterocyte. In cases of pulmonary involvement, vegetables, and other food that may be eaten
the parasite may be recovered from the sputum, raw. Unpasteurized milk, freshly pressed apple
although transbronchial and broncheo-alveolar cider, potato salad, and sausages were found
lavage can yield a better result. sources of infection. Nosocomial infections have
Indirect fluorescent antibody, enzyme also been reported among health workers caring
immunoassay, and DNA probes specific for C. for AIDS patients.
hominis have been developed. Acid-fast staining In developing countries, prevalence
is probably the quickest and cheapest method ranged from 3 to 20%. The prevalence in the
of diagnosis. Philippines has been reported to be low at 2.6%.
62 MedICal parasItology In the phIlIppInes

A study done in San Lazaro Hospital attempted gametes. The microgametes fertilize the
to describe Cryptosporidium among diarrheic macrogametes to produce oocysts, which are
patients and reported a prevalence of 8.5%, passed out with feces when the host cells are
while a study done in the Philippine General sloughed off from the intestinal wall. The
Hospital on diarrheic patients had a much lower oocysts undergo complete sporulation within
prevalence at 1.7%. 7 to 12 days in a warm environment.
It is assumed that the oocyst is the infective
Prevention and Control
stage and when ingested, the sporozoites are
Water-borne transmission is the most released and enter intestinal cells to go through
common source of cr yptosporidiosis. schizogony and gametogony. The different
Chlorination does not affect the parasite. The developmental stages of the parasite may be
synergistic effect of multiple disinfectants and found in the intestinal tissue (Figure 2.9).
combined water treatment processes may reduce
Pathogenesis and Clinical Manifestations
C. hominis oocysts in drinking water. Natural
water and swimming pool water should not be Initial symptoms include malaise and low
swallowed. Contamination of drinking water by grade fever, which may occur 12 to 24 hours
human and animal feces should be prevented. after exposure. Chronic and intermittent watery
diarrhea occurs early in the infection and may
Cyclospora cayetanensis alternate with constipation. The diarrhea
may continue for 6 to 7 weeks with six or
When first associated with diarrhea,
more stools per day. Other symptoms such as
this organism was thought to be a
fatigue, anorexia, weight loss, nausea, vomiting,
m ember of cyan o ba ct er i a b e cau s e it
abdominal pain, flatulence, bloating, and
showed photosynthesizing organelles and
dyspnea may develop. D-xylose malabsorption
autofluorescing particles characteristic of the
has been found to develop in some of the
blue green algae.
patients. Infections are usually self-limiting
Parasite Biology and immunity may result with repeated
infections. No death has been associated with
Cyclospora cayetanensis was originally cyclosporidiosis.
called a cyanobacterium-like body (CLB)
but upon careful study, it was found to be Diagnosis
a coccidian parasite. Similar to the other
Direct microscopic examination of fecal
intestinal coccidians, the life cycle begins with
smears under high magnification (400x)
the ingestion of sporulated oocyst, which
is recommended. Various concentration
contains two sporocysts with two sporozoites
techniques and acid-fast staining (Kinyoun’s
each. The released sporozoites invade the
stain) are also useful. Oocysts are auto-
epithelial cells of the small intestines, although
fluorescent and under fluorescent microscopy,
the site of predilection was found to be the
they appear as blue or green circles depending
jejunum. Multiple fissions of these sporozoites
on the filter (365-450 DM). This technique
take place inside the cells to produce meronts,
is useful for screening. Safranin staining
which contain 8 to 12 merozoites during the
and microwave heating are also helpful. A
first generation, and only four merozoites in
polymerase chain reaction (PCR) technique has
the second generation. Some of the merozoites
been developed to differentiate Cyclospora from
develop into male (micro) and female (macro)
closely related Eimeria species.
Chapter 2: protozoan Infections 63

Figure 2.9. Life cycle of Cyclospora cayetanensis


(Accessed from www.dpd.cdc.gov/dpdx)
64 MedICal parasItology In the phIlIppInes

Treatment followed to prevent the infection. Only water


that has been subjected to adequate treatment
The disease is self-limiting and treatment
procedures should be consumed. In most
is not necessary if the symptoms are mild. If
endemic areas, boiling water seems to be the best
pharmacologic treatment is warranted, the only
method since chlorination is not effective. Fruits
effective drug is trimethoprim-sulfamethoxazole
and vegetables should be washed with clean
160/800 mg twice daily for 7 days. There is
water, but it would be prudent to avoid eating
no alternate treatment if patients are unable to
fruits and vegetables that have been exposed
tolerate sulfamethoxazole. Oocysts disappear
to natural untreated water. In Guatemala, it
from the stools a few days after treatment.
was believed that raspberries were exposed to
However, recurrence of symptoms was noted
oocysts in places where creek water was used
in about 40% of patients within 1 to 3 months
to dilute insecticides sprayed on the plants.
post treatment.
Similarly, in Nepal it is believed that cabbage
Epidemiology became contaminated when watered with raw
irrigation water.
Cyclosporidiosis has been described in
many countries, with epidemics reported in Cystoisospora belli
Nepal, Peru, Haiti, and the United States.
Infections were reported to appear in Nepal in This is the causative agent of a medical
late May and June and continued until October condition affecting the small bowel called
to November, the rainy season. Most cases in cystoisosporiasis. The other known species
Nepal were reported in expatriates and tourists, Isospora hominis is now taxonomically grouped
and more recently in Nepali children and adults. under the genus Sarcocystis.
In Peru, infections are commonly reported in
Parasite Biology
children, while in Haiti, infections affect more
homosexual males. Epidemics involving over The sporulated oocyst contains two
1,000 persons were reported in the United sporocysts each containing four sporozoites
States in 1996 and 1997. Raspberries imported (infective stage). When ingested via contaminated
from Guatemala were incriminated in the water or food, the sporozoites excyst in the small
infections in the United States. Leafy vegetables intestine releasing sporozoites, which penetrate
have been found to contain oocysts in Peru and the epithelial cells, thus starting the asexual
Nepal, while lettuce and basil-pesto salad has stage or the schizogonic phase of the life cycle
been incriminated in other cases in the United (Figure 2.10). The sporozoites develop in the
States. Contaminated water is thought to be the epithelial cell to form a schizont, which ruptures
main source of infection. No animal reservoirs the host epithelial cell liberating merozoites into
have been found and, therefore, cyclosporidiosis the lumen. These merozoites will then infect
is presently considered mainly as a human new epithelial cells and the process of asexual
disease. In the Philippines, a study of diarrheic reproduction in the intestine continues. This
stools from children in 2005 at the College of process may continue for weeks or months.
Public Health, University of the Philippines Some of the merozoites undergo gametogony
Manila, revealed a prevalence of 3.1% using to produce macrogametes and microgametes
safranin staining heated in a microwave. (sexual stages), which fuse to form a zygote that
eventually matures to form an unsporulated
Prevention and Control
oocyst. Sporulation usually occurs within 48
Since the direct source of C. cayetanensis hours after passage with the stool.
is unknown, good sanitary practices should be
Chapter 2: protozoan Infections 65

Figure 2.10. Life cycle of Cystoisospora belli


(Accessed from www.dpd.cdc.gov/dpdx)
66 MedICal parasItology In the phIlIppInes

Pathogenesis and Clinical Manifestations Other concentration techniques that can also
be used include zinc sulfate and sugar flotation.
Among the immunocompetent, infection is
Oocysts are thin walled, transparent, and ovoid
generally asymptomatic or may present as a self-
in shape. They appear as translucent, oval
limiting gastroenteritis. However, in more severe
structures measuring 20 to 33 µm by 10 to
infections, severe diarrhea and fat malabsorption
19 µm. Alternatively, oocysts can be seen in a
can occur. Symptoms include low-grade fever,
fecal smear stained by a modified Ziehl-Neelsen
anorexia, vomiting, general body malaise,
method, where they stain granular red color
anorexia, weight loss, and flatulence. Stools
against a green background. Phenol-auramine,
usually contain undigested food, mucus, and
as well as iodine staining of the specimen
Charcot-Leyden crystals.
can help visualize the organism. Acid-fast
Infection in immunocompromised
stain, such as Kinyoun’s stain or an auramine-
individuals ranges from a self-limiting enteritis
rhodamine stain, is also useful. A considerable
to severe diarrheal illness resembling that of
amount of stool may have to be examined
cryptosporidiosis, giardiasis or cyclosporiasis.
because oocysts in the samples are often few in
Mucosal bowel biopsy may reveal flattened
number. Charcot-Leyden crystals may be seen
mucosa and damaged villi. Infiltration of the
in the stool specimen. In blood examination,
lamina propria with lymphocytes, plasma cells,
peripheral eosinophilia is common. String
and eosinophils has been reported. However,
capsule (Enterotest®) and duodenal aspirate
the mechanism by which the parasite produces
examinations may be of value. Molecular based
these lesions is still not clear.
techniques may prove useful as an additional
Diagnosis diagnostic tool.
The oocysts of C. belli may be detected Treatment
in the feces by direct microscopy or formalin-
Asymptomatic infections may be
ether/ethyl acetate concentration (Plate 2.13).
managed with bed rest and a bland diet,
while symptomatic infections, such as those
occurring in AIDS patients, can be treated with
trimethoprim-sulfamethoxazole 160/800 mg
four times per day for 10 days, then two times
per day for 3 weeks. Combination therapy with
pyrimethamine and sulfadiazine for 7 weeks has
also been used successfully.
Epidemiology

Unlike the other coccidians, humans are


the only known hosts of C. belli, which has
a worldwide distribution. It is however more
common in tropical and subtropical countries
with poor sanitary conditions. The actual
incidence of cystoisosporiasis is not known but
C. belli has been tagged as the causative agent
Plate 2.13. Immature oocyst of Cystoisospora of diarrheal episodes in day care centers and
belli recovered from stool sample,
showing a single sporoblast
mental institutions. The disease is common
(Accessed from www.dpd.cdc.gov/dpdx) among patients with AIDS. In Africa, 2 to 3%
Chapter 2: protozoan Infections 67

of those with AIDS were infected; in South Microsporidia, Isospora and Cyclospora. Ann
America, 10%, and in Haiti and Africa, a range Intern Med. 1996;124:429–441.
of 7 to 20% was observed. The disease has also He y w o r t h M F. Pa r a s i t i c d i s e a s e s i n
been reported among those with lymphoma, immunocompromised hosts,
leukemia, and organ transplants. Considered cr yptosporidiosis, isosporiasis and
endemic are the following: Africa, Australia, strongyloidiasis. Gastroenterol Clin North
the Caribbean Islands, Latin America, and Am. 1996;25:691–707.
Southeast Asia. Cystoisosporiasis has been Hoepelman IM. Human cryptosporidiosis. Int
reported in both adults and children, but severe J STD AIDS. 1996;7(suppl)l:28–33.
diarrhea is common among infants. Both sexes Jueco NL, Belizario VY, Jr., de Leon WU,
were found susceptible to infection. Tan-Liu N, Bravo LC, Gregorio GV.
Cryptosporidiosis among selected patients
Prevention and Control
in the Philippine General Hospital. Acta
Cystoisosporiasis can be prevented by Med Philipp. 1991;27:244–247.
following good sanitary practices, thorough Lindsay DS, Dubey JP, Blagburn BL. Biology
washing and cooking food, and drinking safe of Isospora spp. from humans, non human
water. primates and domestic animals. Clin
Microbiol Rev. 1997;10:19–34.
References
MacKenzie WR, Hoxie NJ, Proctor ME,
A c k e r s J P. G u t C o c c i d i a — I s o s p o ra , Gradus MS, Blair KA, Peterson DE, et
Cryptosporidium, Cyclospora and Sarcocystis. al. A massive outbreak in Milwaukee of
Semin Gastrointest Dis. 1997;8(1):33–44. Cryptosporidium infection transmitted
Brennan MK, MacPherson DW, Palmer J, through the Public water supply. N Engl J
Keystone JS. Cyclosporiasis: a new cause of Med. 1994;331:161.
diarrhea. CMAJ. 1996;155(9):1293–1296. Marshall MM, Naumovitz D, Ortega, Sterling
Cross JH, Serchand JB, Sharma P, Escheverria CR. Waterborne protozoan pathogens.
P. Cyclosporiasis at the Kanti Children’s Clin Microbiol Rev. 1997;10:67–85.
hospital in Kathmandu, Nepal: a cursory Millard PS, Gensheimer KF, Addis DG, Sosin
survey. J Trop Med Parasitol. 1997;20:30– DM, Beckett GA, Houck-Jankoski A,
32. et al. An outbreak of cryptosporidiosis
Duszynski D, Upton S, Couch L. The coccidia from fresh-pressed apple cider. JAMA.
of the world 1995, a compilation of the 1994;272:1592.
national science foundation a database Orenstein JM. Isosporiasis. In: Connor D.
of known species of coccidian [Internet]. et al, editors. Pathology of Infectious
New Mexico and Kansas: University of Diseases. Connecticut: Appleton and
New Mexico and Kansas State University; Lange, Norwalk; 1997. p. 1185–90.
1995 [cited 2009 Mar 1]. Available from Ortega YR. Cyclospora species a new protozoan
http://www.k-state.edu/parasitology/ pathogen of humans. N Eng J Med. 1993;
worldcoccidia/. 328:1308–1312.
Fayer R. Cryptosporidium and cryptosporidiosis. Ortega YR, Sanchez R. Updates on Cyclospora
Florida: CRR Press, Boca Raton; 1997. cayetanensis- a food and water-borne parasite.
p. 251. Clin Microbiol Rev. 2010;23(1):218–234.
Goodgame RW. Understanding intestinal Ro s e J B . E n v i r o n m e n t a l e c o l o g y o f
spore forming protozoa: Cryptosporidia, Cryptosporidium and Public Health
68 MedICal parasItology In the phIlIppInes

I m p l i c a t i o n s . A n n u R e v Pu b l i c S m i t h H V, R o s e J B . Wa t e r b o r n e
Health.1997;18:135–161. cryptosporidiosis: current status. Parasitol
Serchand JB, Cross JH, Jimba M, Serchand M, Today. 1998; 14:14–22.
Shresta MP. Study of Cyclosporacayetanensis Soave R. Cyclospora an overview. Clin Infect Dis.
in healthcare facilities, sewage water 1996;23:429–437.
and green leafy vegetables in Nepal. Stuart RL. Cryptosporidiosis in patients with
Southeast Asian J Trop Med Public Health. AIDS. Int J STD AIDS. 1997;8:339–341.
1999;30:58–63. Taylor AP, Davis LJ, Soave J. Cyclospora. Curr
Clin Top Infect Dis. 1997;17:256–261.
Chapter 2: protozoan Infections 69

Toxoplasma gondii

T oxoplasma gondii is a coccidian that belongs


to the Phylum Apicomplexa. It is a parasite
that has a worldwide distribution and that
into a tachyzoite (Plate 2.14). Tachyzoites are
found during the initial and acute stage of the
infection, but as host immunity to the parasite
infects humans and many species of animals. is developed, the fast multiplying tachyzoites
give rise to slow multiplying bradyzoites that
Parasite Biology
form cysts. Only these two stages are present
The infective stages include the tachyzoite, in humans and other animal intermediate
the bradyzoite, and the oocyst. The complete hosts. Asexual multiplication is by a variation
life cycle occurs only in the members of the cat of binary fission called endodyogeny. This is
family (Felidae), which serve as definitive hosts. characterized by the formation of the plasma
It follows a typical coccidian life cycle consisting membrane by the two new daughter parasites,
of schizogony, gametogony, and sporogony in even before the division of the nucleus. Cells
the intestinal epithelium. The extraintestinal in which endodyogeny occur eventually burst,
stages are the asexual stages: tachyzoites and thus liberating trophozoites that invade other
bradyzoites. cells. It is possible that tachyzoites can be
In the intestinal epithelium of the cat, transferred from one person to another by
merozoites multiply (schizogony) and then granulocyte blood transfusion. Tachyzoites
differentiate into microgametocytes and can be transferred from the newly infected
macrogametocytes (gametogony). Fertilization mother to the fetus during the first trimester
of the macrogamete by the microgamete gives of pregnancy by passing through the placental
rise to an oocyst. The oocyst is ovoidal in shape, barrier. Tachyzoites and bradyzoites can be
has a thin wall, and measures 10 to 13 µm by transferred by organ transplant especially bone
9 to 11 µm. marrow, and bradyzoites can be acquired by
These oocysts are passed out with the feces eating meat of infected animals.
of the cat in the unsporulated stage. These can The trophozoite measures 4 to 8 µm
be ingested together with contaminated food in length, 2 to 3 µm width. It is crescent-
or water by another host. The oocysts complete shaped with a pointed anterior and a rounded
sporulation within three to four days. Inside posterior end. Organelles, such as rhoptries
the mature oocyst, two sporocysts are formed, and micronemes, which are associated with
each having four sporozoites. When the mature cell penetration, are found in a short conoid
oocyst reaches the intestine of the new host, it on the anterior end. A spherical nucleus is
excysts and releases four sporozoites which can found in the posterior end. In the infected
penetrate the lamina propria. The parasites macrophage, the parasites prevent the fusion of
gain entry to the lymphatics then spread to the the parasitophorous vacuole that contains the
different organs, tissues, and fluids of the body parasites, with the lysosome and are, thus, not
(Figure 2.11). killed by the lysozyme. Pseudocysts containing
Toxoplasma is an intracellular parasite, proliferating tachyzoites are seen in tissue
which infects different kinds of nucleated cells sections taken from patients suffering from
including macrophages. Following the entry acute infection. These do not have well-formed
of the sporozoite into a new cell, it transforms walls unlike cysts containing many bradyzoites
70 MedICal parasItology In the phIlIppInes

Figure 2.11. Life cycle of Toxoplasma gondii


(Accessed from www.dpd.cdc.gov/dpdx)

that are seen during chronic infections. Cysts


are found in muscles and in the central nervous
system.
Pathogenesis and Clinical Manifestations

Toxoplasmosis is commonly asymptomatic


as long as the immune system of the patient is
functioning well. Many surveys have shown
the presence of antibodies in a large portion
of the population, although the proportion of
patients exhibiting characteristic symptoms of
Plate 2.14. Toxoplasma tachyzoites
toxoplasmosis is very low. Once stimulated,
(Courtesy of the Department of Parasitology, the immune system quickly responds to the
UP-CPH) parasites, which, in turn, adapt by transforming
Chapter 2: protozoan Infections 71

into bradyzoites that are protected by a cyst antibodies against T. gondii. A seroconversion
wall and proliferate at a slower rate. Cysts to a positive titer or a four-fold increase in titers
can be found in the brain, skeletal and heart is indicative of an infection. The Sabin-Feldman
muscles, and retina. Clinical manifestations methylene blue dye test is very sensitive and
become apparent when the immune system specific but it requires the maintenance of
is suppressed as in old age, drug-induced live organisms in the laboratory. High titers
immunosuppression after organ transplantation, (>1,024), although usually indicating an acute
or in the case of AIDS. More often, symptoms infection, may also be seen in chronic cases,
appear when there is relapse of chronic hence the need for IgM antibody detection
infections as a result of a suppressed immune through either the IgM indirect fluorescent
system rather than as a response to an acute antibody technique or through a double
infection. Among the immunocompromised sandwich IgM enzyme immunoassay. Handling
patients, the most common manifestation is of live trophozoites may result in accidental
encephalitis. Myocarditis and focal pneumonia infection of the laboratory personnel. Other tests
have also been reported. It is also possible are the indirect hemagglutination test, indirect
that the immunosuppressed patient acquires fluorescent antibody test, and enzyme-linked
the infection from blood transfusion or immunosorbent assay. Latex agglutination test
organ transplantation. Clinical manifestations is also available. Differentiating pre-existing
include retinochoroiditis, lymphoreticular antibody from passively transferred antibody
hyperplasia with enlargement of the posterior from the mother or antibody related to illness
cervical lymph node, hepatitis, splenomegaly, is important in the assessment of serological
pneumonia, extramedullary hematopoiesis, and test results.
failure to gain weight. Better diagnostic assays are being developed
Stillbirth and abortion may result when because toxoplasmosis has been recognized
mothers acquire the infection during the first as an important disease associated with
trimester of pregnancy. Babies may exhibit AIDS. Polymerase chain reaction (PCR) has
clinical manifestations like chorioretinitis, been successfully used in the diagnosis of
epileptic seizures, jaundice, hydrocephaly, and toxoplasmosis using samples taken from the
microcephaly. Death of the infected newborn patient, which include serum, amniotic fluid,
babies is usually due to anemia with pneumonia. cerebrospinal fluid, and broncheoalveolar
There are cases when clinical manifestations lavage, especially in cases where there is very
may not be apparent during the neonatal little amount of specimen available.
period, but will appear later in childhood. Most
Treatment
babies will harbor the infection and grow up
without any clinical manifestation until such Treatment consists of pyrimethamine
time later in life when their immune system is (25-100 mg daily) and sulfadiazine (1-1.5 g
suppressed and there is reactivation of chronic four times daily) used in combination for one
toxoplasmosis. month. These drugs keep the Toxoplasma under
control but do not kill it. Since pyrimethamine
Diagnosis
can lower blood counts in most people, it
Identification of the parasite can be done should be given together with leucovorin (folic
through examination of tissue imprints stained acid). Sulfadiazine may cause serious allergic
with Giemsa. Tissue sections can be processed reactions like fever and rash, but it can be
and stained with hematoxylin and eosin. substituted with clindamycin. Spiramycin,
Serodiagnostic methods are used to detect azithromycin, clarithromycin, dapsone, and
72 MedICal parasItology In the phIlIppInes

atovaquone may also be used. Corticosteroids References


are sometimes given to prevent occurrence of
Cross JH, Basaca-Sevilla V. Biomedical surveys
hypersensitivity reactions. Prophylaxis with
in the Philippines. Manila (Philippines):
trimethoprim-sulfamethoxazole may be given
US Naval Medical Research Unit No. 2;
for the immunocompromised.
1984.
Epidemiology Eduardo SL. Food-borne zoonoses in the
Philippines. Southeast Asian J Trop Med
Toxoplasmosis is endemic worldwide in
Public Health. 1991;22:16–22.
humans and in domestic and wild animals as
Frenkel JK, Hassanein KM. Transmission of
well. Disease due to this parasitic infection is not
Toxoplasma gondii in Panama: a five-year
manifested except in cases of immune deficiency
prospective cohort. Study of children, cats,
or suppression. Determination of the prevalence
rodents and soil. Am J Trop Med Hyg.
of infection is based on serodiagnostic tests,
1995;53:458.
although these tests are not readily available
Jackson MH, Hutchison WM. The prevalence
in the Philippines due perhaps to a lack of
and source of Toxoplasma infection in the
demand since clinical toxoplasmosis is not
environment. Adv Parasitol. 1989;28:55–
common. According to surveys by Cross and
105.
Basaca-Sevilla, only 2.4% of the population is
Nantulya VM. TrypTect CIATT—a card
seropositive for Toxoplasma gondii. Pigs and rats,
indirect agglutination trypanosomiasis test
however, have a higher prevalence of positive
for diagnosis of Trypanosoma gambiense and
titers for Toxoplasma antibodies at 19% and
T. rhodesiense infections. Trans R Soc Trop
8.1%, respectively.
Med Hyg. 1997;91:55l–553.
Prevention and Control Neva FA, Brown HW. Basic clinical parasitology.
6th ed. Connecticut: Appleton & Lange;
Fo o d s h o u l d b e p r o t e c t e d f r o m
1944.
contamination with cat feces. Meat and eggs
Roberts LS, Janovy J. Foundations of
should be well cooked. Unpasteurized milk
parasitology. 5th ed. Dubuque: Wm. C.
should be avoided. Pregnant women should
Brown Publishers; 1996.
avoid contact with cats. Laboratory workers
World Health Organization. WHO Fact
should be very careful in handling the parasite.
Sheet no. 116. Geneva: World Health
Organization; 1996.
Chapter 2: protozoan Infections 73

Sarcocystis spp.
Alice Alma C. Bungay, Raezelle Nadine T. Ciro

S arcocystis is a genus of intracellular protozoa


reported to infect humans and animals
worldwide. Infection with this parasite is known
environment suitable for parasite growth and
development.
Sporulated oocysts and individual
as sarcosporidiosis or sarcocystosis. Species sporocysts can be passed out in the feces of an
belonging to this genus infect a wide variety of infected definitive host. The sporulated oocyst
animals such as birds, reptiles, and mammals. undergoes sporogony creating two sporocysts.
While majority of the species infect mammals, Once sporogony is complete, the oocyst itself
about a dozen are known to infect snakes. undergoes lysis, releasing the sporocysts into
This parasite was first reported in 1843 by the environment. Sporocysts of most species
Miescher as white threadlike cysts in striated measure 15 to 19 µm by 8 to 10 µm, and contain
muscles of a house mouse. It was simply referred four sporozoites and a discrete refractile residual
to as Miescher’s tubules until 1899, when the body. Sporocysts are capable of surviving on
name Sarcocystis miescheriana was proposed to the ground and infecting intermediate hosts
identify the said parasite. Since its discovery, it (Figure 2.12).
has been debated whether Sarcocystis spp. were After oocysts and/or sporocysts are ingested
protozoa or fungi. The debate was resolved by a susceptible intermediate host (usually
only in 1967 when bradyzoites in the sarcocysts cows or pigs), the sporocysts pass to the small
were studied under the electron microscope and intestine. The plates forming the sporocyst wall
were seen to possess organelles found in other separate, releasing the four sporozoites into
apicomplexan protozoa such as Toxoplasma and the intermediate host’s body. The sporozoites
Eimeria. migrate through the gut epithelium and
There are about 130 recognized species eventually enter the endothelial cells in small
under Sarcocystis including S. hominis and S. arteries where they undergo the first two
suihominis. Humans serve as definitive hosts for generations of asexual reproduction (called
the two species, but occasionally, humans can schizogony or merogony). These cycles result
act as intermediate hosts. There is an ongoing in the development of meronts. This stage lasts
revision of the taxonomy of this genus, and it is about 15 to 16 days after ingestion of sporocysts.
possible that all the currently recognized species Merozoites emerge from the second generation
may be fewer or may in fact be a single species meronts and enter the mononucleate cells
that can infect multiple hosts. where they develop. Subsequent generations of
merozoites develop in the direction of blood
Parasite Biology
flow to arterioles, capillaries, venules, and
Sarcocystis can take several forms. The veins throughout the body. The third asexual
simplest form is called a zoite. It is a banana- generation appears as multinucleate schizonts
shaped cell, with a pointed anterior end, also in capillaries throughout the body. Merozoites
known as the apical complex, which possesses from this generation form metrocytes and encyst
micronemes, micropores, and rhoptries, in the muscles, initiating sarcocyst formation.
and believed to be associated with host cell Sarcocysts begin as unicellular bodies
penetration and creation of an intracellular containing a single metrocyte. Through
74 MedICal parasItology In the phIlIppInes

Figure 2.12. Life cycle of Sarcocystis spp.


(Accessed from www.dpd.cdc.gov/dpdx)
Chapter 2: protozoan Infections 75

repeated asexual multiplication, numerous anorexia, nausea, abdominal pain, distension,


metrocytes accumulate and the sarcocyst diarrhea, vomiting, dyspnea, and tachycardia.
increases in size. As sarcocysts mature, the All symptoms were transient and lasted about
small, rounded, non-infectious metrocytes give 36 hours.
rise to infectious, crescent-shaped bodies called Sarcocystosis has also been associated with
bradyzoites. About two and a half months after acute fever, myalgias, bronchospasm, pruritic
infection, sarcocysts are already mature and are rashes, lymphadenopathy, subcutaneous
able to infect the definitive host. nodules with concurrent eosinophilia, elevated
Humans, as well as other definitive hosts, erythrocyte sedimentation rate, and elevated
are infected by consumption of uncooked or creatine kinase levels. Symptoms may last as
undercooked meat of intermediate host that long as 5 years. Segmental necrotizing enteritis
contains sarcocysts. Once the intermediate has also been reported in one study.
host is eaten by a definitive host such as dog
Diagnosis
or human, the parasite undergoes sexual
reproduction within the intestines. After Presumptive diagnosis of human intestinal
sarcocysts are ingested and the wall is digested, sarcocystosis is based on symptoms manifested
bradyzoites become motile. Active bradyzoites by infected individuals and a history of recent
enter intestinal cells and change into the consumption of raw or undercooked meat.
male and female forms, microgamonts and Identification of sporocysts in feces may
macrogamonts, respectively. Fusion of a require several stool examinations done on
macrogamont and a microgamont creates a separate days during the infection. Sporocysts
fertilized cell called a zygote, which develops of S. hominis are first excreted 14 to 18 days
into an oocyst (containing two sporocysts). after ingesting beef, and those of S. suihominis
The oocyst is passed through the feces of the are excreted 11 to 13 days after ingesting
definitive host. Most definitive hosts do not pork. A fecal flotation wet mount is usually
show any clinical signs or symptoms. done to visualize sporocysts using bright-field
More recently, a second life cycle has been microscopy. Flotation methods based on
described whereby carnivores and omnivores high-density solutions incorporating sodium
pass the infectious stages in their feces. Ingestion chloride, cesium chloride, zinc sulfate, sucrose,
of this contaminated material may lead to Percoll, Ficoll-Hypaque, and other density
successful infection. gradient media are preferred over formalin-
ether/ethyl acetate and other sedimentation
Pathology and Clinical Manifestations
methods. Species cannot be distinguished
The pathology is of two types: a rare from one another solely by microscopy because
invasive form that presents with vasculitis and sporocysts of different species overlap in size
myositis, and an intestinal form that presents and shape.
with nausea, abdominal pain, and diarrhea. Definitive diagnosis can be made through
While normally mild and lasting under 48 biopsy of an infected muscle. Sarcocysts of S.
hours, the intestinal form may occasionally be hominis are microscopic in muscles of cattle,
severe or even life threatening. The invasive whereas those of S. suihominis are macroscopic
form may involve a wide variety of tissues in muscles of swine. Sarcocysts are identifiable
including lymph nodes, muscles, and the larynx. with hematoxylin and eosin stain. Confirmatory
In studies where volunteers ingested staining with the periodic acid-Schiff (PAS) can
infected beef, symptoms appeared 3 to 6 be performed as the walls stain positively. The
hours after eating. These symptoms included walls of the sarcocyst may be used in species
76 MedICal parasItology In the phIlIppInes

diagnosis. Currently, 24 wall types have been America, China, India, Tibet, and Southeast
identified in 62 species. S. hominis and S. Asia.
suihominis both have walls of type 10. The wall Of fecal specimens examined from children
of S. hominis is up to 6 µm thick and appears in Poland and Germany, 10.4% and 7.3% were
radially striated from villar protrusions that are found positive, respectively. In Tibet, Sarcocystis
up to 7 µm long. The wall of S. suihominis is was detected in 42.9% of beef specimens
4 to 9 µm thick, with villar protrusions up to examined from the marketplace, and S. hominis
13 µm long. and S. suihominis were found in stool samples
Recently, polymerase chain reaction of 21.8% and 7% of 926 persons, respectively.
(PCR) amplification of the 18S rRNA was Stool examination among Thai laborers showed
demonstrated to be useful in distinguishing S. that Sarcocystis infection had a prevalence of
hominis, S. fusiformis, and S. cruzi sarcocysts about 23%; all cases were asymptomatic which
and oocysts. The technique makes possible probably explained the lack of recognition. A
amplification and identification of species- study of 100 human tongues obtained post
specific gene sequences based on DNA extracted mortem in Malaysia revealed an infection rate
from as few as seven excreted sporocysts (the of 21%. There was no sex difference and the age
equivalent of 3 ½ oocysts) from freshly prepared range was 16 to 57 years (mean 37.7 years). A
material, or as few as 50 sporocysts from fecal seroepidemiological survey in West Malaysia
samples that had been stored in potassium found that 19.7% of 243 persons had antibodies
dichromate (K2Cr2O7) for as long as 6 years. for Sarcocystis.
In the Philippines, studies involving
Treatment
the examination of muscle tissues obtained
Because infection is often asymptomatic, from water buffaloes, cattle, pigs, and goats
treatment is rarely required. There have been revealed the presence of S. cruzi in backyard
no published trials so treatment remains cattle (Bos taurus) possessing a type 7 sarcocyst
empirical. Agents that have been used include wall, S. levinei in water buffaloes (Bubalus
albendazole, metronidazole, and co-trimoxazole bubalis) possessing a type 7 sarcocyst wall
for myositis. Corticosteroids have also been used with similarities to S. cruzi, S. miescheriana in
for symptomatic relief. domestic pigs (Sus scrofa domestica) with a type
10 sarcocyst wall, and S. capracanis in domestic
Epidemiology
goats (Capra hircus) with a type 14 sarcocyst
There are very few large-scale population wall. There is a lack of local studies on human
surveys that have been conducted for Sarcocystis sarcocystosis.
in humans. Prevalence data for Sarcocystis Prevention and Control
infections often come from case reports and
findings of physicians, public health workers, Intestinal sarcocystosis can be prevented
and scientists with specific interests. by thoroughly cooking or freezing meat to kill
Human infection is considered rare with bradyzoites in the sarcocysts. Alternatively,
less than 100 published cases of invasive freezing the meat at –5°C for several days
disease (approximately 46 cases reported by will kill the sporocysts. Where contaminated
1990). These figures may represent a gross drinking water is suspected, boiling should be
underestimate of the human burden of disease. considered to ensure disinfection.
Sarcocystosis has been reported in Africa, The administration of anticoccidial
Europe (Germany, Spain, and Poland), the drugs, amprolium and salinomycin, as
United States (California), Central and South chemoprophylactic agents was effective
Chapter 2: protozoan Infections 77

in preventing severe illness and death in Research@DLSU-Manila:_Continuing_


experimentally infected calves and lambs. the_Cycle. 2007;100.
The risk of foodborne zoonoses warrants Croft JC. Nonamebic protozoal enteridities.
prevention and control in food animals. To In: Hoeprich D, Jordan MC, Ronald AR.
avoid infection of food animals, they must be Infectious processes. 5th ed. Philadelphia,
prevented from ingesting the sporocyst stage Pa: Lippincott; 1994. p. 769–74.
from human feces in contaminated water, feeds, Dubey JP, Speer CA, Fayer R. Sarcocystis of
and bedding. If such measures cannot be assured animals and man. Boca Raton, Fla.: CRC
and meat is suspected to harbor cysts, the extent Press, Inc.; 1989.
of infestation must be considered. In heavy and Herenda D, Chambers PG, Ettriqui A,
widespread infestations with visible cysts, the Seneviratna P, da Silva TJ. Manual on meat
whole carcass must be condemned. In lighter inspection for developing countries. Rome,
infestations, those parts of the carcass which are Italy: FAO; 2000.
not affected are passed for human consumption. Leek RG, Fayer R. Experimental Sarcocystis
No vaccines are currently available. ovicanis infection in lambs: salinomycin
Experimentally inoculated pigs appear to chemoprophylaxis and protective
develop a persistent immunity, hence, vaccine immunity. J Parasitol. 1983;69:271–6.
development may be explored. Ohio State University. Sarcocystis spp [Internet].
2010 [cited 2010 Mar 1]. Available from
References
http://www.biosci.ohiostate.edu/parasite/
Bruckner DA, Garcia LS. Diagnostic medical sarcocystis.html
parasitology. UCLA Medical Center, Payer R. Sarcocystis spp. in human infections.
Department of Pathology: Elsevier Science Clin Microbiol Rev. 2004;17(4):894–902.
Publishing Co., Inc; 1988. Xiang Z, Chen X, Yang L, He Y, Jiang R,
Charleston WAG, Pomroy WE. Sarcocystis Rosenthal BM, et al. Non-invasive methods
species: self-teaching manual for veterinary for identifying oocysts of Sarcocystis spp.
parasitology. Massey University: VPPH from definitive hosts. Parasitol Int. 2009;
Publication; 1995. 58(3):293–6.
Claveria FG. Survey of Sarcocystis spp. infection Yu S. [Field survey of Sarcocystis infection in the
in Philippine livestock animals: light Tibet autonomous region]. Zhongguo Yi
microscopic and ultrastructural studies. Xue Ke Xue Yuan Xue Bao. 1991;13:29–
32. Chinese.
78 MedICal parasItology In the phIlIppInes

Other Intestinal Protozoans


Winifreda U. de Leon

Blastocystis hominis Parasite biology

The life cycle is unclear. It has been


B lastocystis hominis is an intestinal protozoan
found in a vast array of animals, including
humans. The classification of Blastocystis has
proposed that the life cycle begins with ingestion
of cysts from contaminated food or water. Upon
ingestion, the cyst possibly develops into other
been a long-standing problem for taxonomists.
forms, which may in turn re-develop into cyst
It was previously classified as yeast under
forms. When excreted with stools, the cysts
the genus Schizosaccharomyces, while other
contaminate the environment and are eventually
taxonomists suggested that it was related to
transmitted to humans and other animals
Blastomyces based on its glistening appearance
through the fecal-oral route, repeating the cycle.
in a wet mount and the absence of any organelle
Because the life cycle is not fully understood,
of locomotion.
validation of this proposed life cycle and the
Correlative light electron microscopy has
mode of transmission needs experimental
since shown that the organism lacks a cell wall. It
confirmation. Multiplication of B. hominis is
possesses nuclei, endoplasmic reticulum, Golgi
by binary fission.
complex, and mitochondrion-like organelles
B. hominis is known to occur in four
that are compatible with protozoan morphology.
morphological forms: (a) vacuolated, (b) ameba-
It is capable of pseudopodial extension and
like, (c) granular, and (d) multiple fission. More
retraction. Moreover, the organism does not
recently, additional cyst and avacuolar forms
grow on fungal culture media. It responds to
have been recognized.
anti-protozoal drugs. Studies with cultured
Vacuolated forms are the most predominant
organisms have shown that reproduction
forms in fecal specimens. These are spherical in
is asexual, either through binary fission or
shape, measuring 5 to 10 µm in diameter. A
sporulation under strict anaerobic conditions.
large central vacuole pushes the cytoplasm and
Optimal growth is at 37°C in the presence of
the four nuclei to the periphery of the cell.
bacteria. All the above findings supported the
Sometimes, a very thick capsule surrounds
reclassification of B. hominis from a yeast to an
the vacuolated forms. The prominent central
emerging human protozoan parasite.
vacuole has been found to be a reproductive
There are new research findings on the
organelle. The vacuolar forms are considered
taxonomic classification of B. hominis. In
to be the main type of Blastocystis that cause
1996, Silberman et al. completed a study of
diarrhea.
the small subunit rRNA (SSUrRNA) gene
Ameba-like forms, usually measuring
of the organism, and the results showed
between 2.5 to 8 µm, are occasionally observed
that it belongs to an informal group called
in stool samples. They exhibit active extension
Stramenophiles, which is a recently recognized
and retraction of pseudopodia. The nuclear
group of microscopic parasites. This includes
chromatin, when visible, characteristically
heterogenous protists like brown algae, diatoms,
shows peripheral clumping. The amebic form
and water molds, to name a few.
appears to be an intermediate stage between
Chapter 2: protozoan Infections 79

the vacuolar form and the precystic form, as flatulence, mild to moderate diarrhea without
this stage allows the parasite to ingest bacteria fecal leukocytes or blood, nausea, vomiting, low
in order to enhance encystment. Studies grade fever, and malaise. Symptoms usually last
of Tan and Suresh have revealed that the about 3 to 10 days, but may sometimes persist
ameboid forms predominated in isolates from for weeks or months.
symptomatic cases. It has been found that in subjects suffering
Granular forms are multinucleated and from immunosuppression, Blastocystis showed
are mainly observed from old cultures. The a significant association with gastrointestinal
diameter of the cell varies from 10 to 60 µm. symptoms. Other studies have also provided
The granular contents develop into daughter evidence of changes in the cellular immune
cells of the ameba-form when the cell ruptures. function of infected individuals.
Multiple fission forms arise from vacuolated
Diagnosis
forms. It is believed that these multiple fission
forms produce many vacuolated forms. Specific diagnosis based on clinical
The size of the resistant cystic form is presentation alone may prove difficult, because
about 3 to 10 µm in diameter, and has one or the spectrum of symptoms is seen in other
two nuclei. It has a very prominent and thick, intestinal infections. Laboratory detection of
osmophilic, electron dense wall. It appears the organism from stool is needed to confirm
as a sharply demarcated polymorphic, but the diagnosis. Multiple stool samples should
mostly oval or circular, dense body surrounded be collected from patients showing clinical
by a loose outer membranous layer. This signs and symptoms. Microscopic examination
membranous layer seen in phase contrast using direct fecal smear is useful, but sensitivity
microscopy corresponds to the fibrillar layer is increased when concentration techniques
described around the cyst at the ultrastructural are used. Hematoxylin or trichrome staining
level, and is the easiest diagnostic feature to offers a very convenient and easy method to
identify. differentiate the various stages of Blastocystis.
It is postulated that the thick-walled cyst Leukocytes are usually seen in fecal smears and
may be responsible for external transmission, stool eosinophilia may also be observed. The
while those cysts with thin walls may be the organism can be cultured using the Boeck and
cause of reinfection within a host’s intestinal Drbohlav’s or the Nelson and Jones media.
tract.
Treatment
Pathogenesis and Clinical Manifestations
Blastocystis is difficult to eradicate. It hides
Infection with B. hominis is called in the intestinal mucus, as well as sticks and
blastocystosis. B. hominis as a cause of holds on to intestinal membranes. The drug of
gastrointestinal pathology is controversial. choice is metronidazole given orally, 750 mg
Several studies have shown that the presence three times daily for 10 days (Pediatric dose:
of the parasite in a majority of patients was not 35-50 mg/kg/day in three doses for 5 days)
associated with symptoms; or, it was found or iodoquinol given at 650 mg three times
with other organisms that were more likely to daily for 20 days. However, there have been
be the cause of the symptoms. However, other reported cases of resistance. Trimethroprim-
studies have concluded that the presence of sulfamethoxazole (TMP-SMX) has also been
Blastocystis in large numbers produces a wide found to be highly effective against Blastocystis.
variety of intestinal disorders, such as abdominal Nitazoxanide has been clinically tested on
cramps, irritable bowel syndrome, bloating, patients with blastocystosis, and was found to
80 MedICal parasItology In the phIlIppInes

resolve symptoms in 86% of patients after 3 Blastocystis similar to those found in humans.
days of administration. Evidence has also shown that Blastocystis is
present in house lizards and cockroaches,
Epidemiology
raising the possibility that food and water
Blastocystis hominis has been reported contaminated by fecal droppings of these “home
virtually worldwide, with infections occurring visitors” may transmit Blastocystis.
most commonly in tropical, subtropical, and In the Philippines, studies of 32
developing countries. Studies from developed morphologically similar isolates from different
countries have reported approximately 1.5 to hosts: 12 from humans, 12 from pigs, and 8
17.9% overall prevalence of B. hominis. All from chickens, using the restriction fragment
ages are affected, but symptomatic cases are length polymorphism (RFLP) analysis of small
more often found in children and in those with subunit rDNA (SSUrDNA), have shown
weakened immune systems. A prevalence of up extensive genomic polymorphism.
to 11.6% was reported from Stanford University
Prevention and Control
Hospital. Prevalence rates of 32.6 % and as high
as 52.3% had been reported from China and Available data on B. hominis indicate that
Malaysia, respectively. the disease can be prevented by consuming safe
Occurrence of the parasite in temperate drinking water. While food has not been fully
countries is generally associated with recent implicated, provisions for sanitary preparation
travel to the tropics and consumption of may be of value in efforts to prevent and
untreated drinking water. This indicates that control this infection. The cysts of B. hominis
infection is possibly through the oral route, can survive up to 19 days in water at normal
and it is more likely to occur in crowded and temperature, and have shown resistance to
unsanitary conditions. Outbreaks of B. hominis chlorine at the standard concentrations.
in day-care centers have been reported in Spain
References
(5.3-10.3%), Brazil (34.7%), and Canada
(13.4%). Avila MS, Garcia MR, Narcelles MV, Serra
In the Philippines, examination of FB, Tejida GM. Prevalence of intestinal
772 stools from consecutive patients at the helminth and protozoan infections among
Department of Parasitology, College of Public food handlers in selected school canteens
Health, University of the Philippines Manila, in Manila [undergraduate special study].
showed a prevalence of 20.7%, sometimes with 2003. Located at: College of Public Health
concomitant infection with other intestinal Library, University of the Philippines
parasites. Studies have also shown prevalence Manila.
rates of 40.6% among food service workers Department of Parasitology. Diagnostic
in a tertiary hospital, and 23.6% among Laboratory Records. 1997. Located at:
food handlers in selected school canteens College of Public Health Library, University
in Manila. Stool surveys conducted by the of the Philippines Manila.
Field Epidemiology Training Program of the Department of Parasitology. Diagnostic
Department of Health in Tapel, Gonzaga, Laboratory Records. 1998. Located at:
Cagayan Valley, and Talavera, Nueva Ecija College of Public Health Library, University
showed prevalence rates of 20% and 44%, of the Philippines Manila.
respectively. Doyle PW, Helgason MM. Epidemiology and
Some animals, like pig-tailed macaques, pathogenicify of Blastocystis hominis. J Clin
chickens, dogs, and ostriches may harbor Microbiol. 1990;28:116–21.
Chapter 2: protozoan Infections 81

Diaczok BJ, Rival J. Diarrhea due to Blastocystis Mclure HM, Strobeft EA, Healy GR.
hominis: an old organism revisited. South 1980 Blastocystis hominis in a pigtailed
Med J. 1987;80(7):931–2. macaque: a potential enteric pathogens
Esparar, DG, Belizario VY. Prevalence of for non-humans primates. Lab Anim Sci.
parasitic infection among food-handlers 1980;30(5):890–4.
in a dietary service of a tertiary hospital Rivera W, Tan MA. Molecular characterization
in Manila. 2003. Located at: College of of Blastocystis isolates in the Philippines
Public Health Library, University of the b y r i b o p r i n t i n g . Pa r a s i t o l R e s .
Philippines Manila. 2005;96(4):253–7.
Garcia LS, Brucknel DA, Clancey MN. Clinical Rivera WL. Phylogenetic analysis of Blastocystis
relevance of Blastocystis hominis. Lancet. isolates from animal and human hosts in the
1984;1:1233–4. Philippines. Vet Parasitol. 2008;156:178–
Guirges SY, Al Waili NS. Blastocystis hominis: 82.
evidence for human pathogenicity and Rossingnol JF, Kabil SM, Said M, Samir H,
effectiveness of metronidazole therapy. Clin Younis AM. Effect of nitazoxanide in
Exp Pharmacol Physiol. 1986;4:333–335. persistent diarrhea and enteritis associated
Haresh H, Suresh K, Khairul A, Saminathan with Blastocystis hominis. Clin Gastroenterol
S. Isolate resistance of Blastocystis hominis Hepatol. 2005;3(10):987–91.
to metronidazole. Trop Med Int Health. Silberman JD, Sogin ML, Leipe DD, Clark CG.
1999;4:274–7. Human parasite finds taxonomic home.
Jiang JB, He, JG. Taxonomic status Blastocystis Nature.1996;380(6573):398.
hominis. Parasitol Today. 1993;9(10):2–3. Tan KS. New insights on classification,
Kain KC, Noble MA, Freeman HJ, Barteluk identification, and clinical relevance
RL. Epidemiology and clinical features of Blastocystis spp. Clin Microbiol Rev.
associated with Blastocystis hominis infection. 2008;21(4):639–65.
Microbiol Infect Dis. 1987;8(4):235–44. Tan TC, Suresh KG. Predominance of Ameboid
Koutsavlis AT, Valiquette L, Allard R, Soto J. forms of Blastocystis hominis in isolates
Blastocystis hominis: a new pathogen in from symptomatic patients. Parasitol Res.
day-care centers? Can Commun Dis Rep. 2005;98(3):189–93.
2001;27:76–84. Valido E, Rivera W. Colony Growth of
Long HY, Handschack A, Konig W. Blastocystis Blastocystis hominis in simplified soft agar
hominis modulates immune responses and medium. Parasitol Res. 2007;101(1):213–
cytokine release in colonic epithelial cells. 7.
Parasitol Res. 2001;87:1029–30. Yoshikawa H, Yoshida K, Nakajima A,
Markell EK, Udkow MP. Blastocystis hominis: Yamanari K, Iwatani S, Kimata I. Fecal-
pathogen or fellow traveler? Am J Trop Med oral transmission of the cyst form of
Hyg. 1986;35(5):1023–6. Blastocystis hominis in rats. Parasitol Res.
Matsamuto Y, Yamada M, Yoshida Y. Light 2004; 94(6):391–6.
microscopical appearance and ultra- Zierdt CH. Blastocystis hominis-past and future.
structure of Blastocystis hominis, an intestinal Clin Microbiol Rev. 1991;4:61.
parasite of man. Zentrabl Bakteriol
Mikrobiol Hyg B. 1986;264(3–4):379–85.
82 MedICal parasItology In the phIlIppInes

Dientamoeba fragilis
Vicente Y. Belizario, Jr., Timothy M. Ting

D ientamoeba fragilis was first discovered by


Wenyon in 1909 but was first described
in the scientific literature by Jepps and Dobell
debris. No cyst stage has been identified. Except
for the absence of a flagellum, this protozoan is
closely related to and resembles Trichomonas.
in 1918. It remains neglected despite evidence D. fragilis lives in the mucosal crypts of
supporting its pathogenicity. It has been the appendix, cecum and the upper colon. The
identified in practically all regions of the world exact life cycle is unknown, although several
in which satisfactory iron-hematoxylin stained assumptions have been made from clinical
films have been carefully examined. data (Figure 2.13). Direct human to human
transmission is probably via the fecal-oral route
Parasite Biology
or via transmission of helminth eggs particularly
On the basis of electron microscopic, that of Enterobius vermicularis. Dientamoeba-
immunologic, and molecular phylogenetic like mononucleated and binucleated forms
findings, this protozoan, which was originally have been observed in the lumen of Enterobius
described as an ameba, is actually a flagellate adults and eggs present in the intestines. More
with only the trophozoite stage known (Plate recently, stools from macaques, gorillas, and
2.15). The organism measures about 7 to 12 µm swine were found to carry D. fragilis, thus
with one or two (rarely three or four) rosette- animal reservoirs may also be potential sources
shaped nuclei. The nuclear membrane does not of human infections.
have peripheral chromatin, and the karyosome
Pathogenesis and Clinical Manifestations
consists of four to six discrete granules. The
cytoplasm may contain vacuoles with ingested Dientamoeba fragilis does not invade
tissues, but its presence in the intestines
produces irritation of the mucosa with secretion
of excess mucus and hypermotility of the
bowel. Infections are usually asymptomatic. In
symptomatic individuals, the onset of infection is
usually accompanied by loss of appetite, colicky
abdominal pain, and intermittent diarrhea with
excess mucus, abdominal tenderness, a bloating
sensation, and flatulence. Another common
symptom, reported in 11% of the patients,
was anal pruritus. This may partially be due
to the co-infection with Enterobius. Peripheral
eosinophilia can be observed in 50% of the
cases. Chronic infection of this organism can
mimic the symptoms of diarrhea-predominant
irritable bowel syndrome (IBS), and some
Plate 2.15. Binucleate forms of trophozoites of experts have suggested ruling out infection with
Dientamoeba fragilis, stained with trichrome this organism first before diagnosing a patient
(Accessed from www.dpd.cdc.gov/dpdx) as having IBS.
Chapter 2: protozoan Infections 83

Figure 2.13. Life cycle of Dientamoeba fragilis


(Accessed from www.dpd.cdc.gov/dpdx)

Diagnosis of the fresh specimen with polyvinyl alcohol


fixative or Schaudinn’s fixative has been found
Diagnosis of this organism is by observation
to be helpful.
of binucleate trophozoites in multiple fixed and
stained fresh stool samples. Fresh stool samples Treatment
are necessary since the trophozoites degenerate
Antimicrobial therapy is followed by
after a few hours of stool passage. Multiple
resolution of symptoms and eradication of
samples increase the sensitivity of detecting
D. fragilis. Treatment is done with iodoquinol
the organism. Unless the laboratory examiner
at 650 mg three times daily for 20 days. The
is aware of the possibility that D. fragilis may be
pediatric dose is 40 mg/kg/day in three doses,
present in the fresh fecal films, the protozoan
also for 20 days. Tetracycline and metronidazole
is easily overlooked. Purged stool specimens
have also been found to be effective.
provide more suitable material for examination
than the average formed stool. Even when Epidemiology
formed, D. fragilis may be misdiagnosed as
The organism has a world-wide distribution
other amebae. This organism is not detected by
with varying infection rates ranging from 0.4 to
stool concentration methods. Prompt fixation
84 MedICal parasItology In the phIlIppInes

as high as 42%. In contrast to many pathogenic population: a preliminary investigation. Vet


protozoa, which have a high prevalence in Parasitol. 2007;145:349–51.
developing countries, high prevalence rates of Girginkardesler N, Kurt O, Kilimcioglu A,
D. fragilis have been reported from developed Ok U. Transmission of Dientamoeba
countries with high sanitation standards. Using fragilis: evaluation of the role of Enterobius
adequate culture techniques, the rates were as vermicularis. Parasitol Int. 2008;57:72–5.
high as 18% in Israel, 36% in Holland, and Johnson E, Windsor J, Clark G. Emerging
41.5% in Germany. from obscurity: biological, clinical, and
diagnostic aspects of Dientamoeba fragilis.
Prevention and Control
Clin Microbiol Rev. 2004;17(3):553–70.
Specific recommendations for prevention Johnson J, Clark C. Cryptic genetic diversity
and control cannot be made until there is more to Dientamoeba fragilis. J Clin Microbiol.
specific information concerning the method of 2000;38(12):4653–4.
transmission. Proper sanitation and disposal of Katz D, Taylor D. Parasitic infections of the
human waste are essential. gastrointestinal tract. Gastroenterol Clin
N. 2001;30(3):797–815.
References
Lagace-Wiens P, Van Caeseele P, Koschik C.
Banik G, Birch D, Stark D, Ellis J. A Dientamoeba fragilis: an emerging role
microscopic description and ultrastructural in intestinal disease. Can Med Assoc J.
characterization of Dientamoeba fragilis: an 2006;175(5):468–9.
emerging cause of human enteric disease. Stark D, Barratt J, Roberts T, Marriott D,
Int J Parasitol. 2012;42:139–53. Harkness J, Ellis J. A review of the clinical
Banik G, Barratt J, Marriott D, Harkness J, presentation of dientamoebiasis. Am J Trop
Ellis J, Stark D. A case-controlled study of Med Hyg. 2010;82(4):614–9.
Dientamoeba fragilis infection in children. Stark D, Philipps O, Peckett D, Munro U,
Parasitol. 2011;138:819–23. Marriott D, Harkness J, Ellis J. Gorillas are
Barratt J, Harkness J, Marriorr D, Ellis J, a host for Dientamoeba fragilis: an update
Stark D. A review of Dientamoeba fragilis on the life cycle and host distribution. Vet
carriage in humans: several reasons why Parasitol. 2008;151:21-6.
this organism should be considered in the Stark D, Beebe N, Marriott D, Ellis J, Harkness
diagnosis of gastrointestinal illness. Gut J. Dientamoeba fragilis as a cause of traveler’s
Microbes. 2011;2(1):3–12. diarrhea: report of seven cases. J Travel
Barratt J, Harkness J, Marriott D, Ellis J, Stark Med. 2007;14(1):72–3.
D. The ambiguous life of Dientamoeba Windsor J, Macfarlane L. Irritable bowel
fragilis: the need to investigate current syndrome: the need to exclude
hypotheses on transmission. Parasitol. Dientamoeba fragilis. Am J Trop Med Hyg.
2011; 138:557–72. 2005;72(5):501.
Crotti D, Sensi M, Crotti S, Grelloni V, Windsor J, Johnson E. Dientantoeba fragilis:
Manuali E. Dientamoeba fragilis in swine The unflagellated human flagellate. Brit J
Biomed Sci. 1999;56(4):293–306.
Chapter 2: protozoan Infections 85

Plasmodium spp.
Vicente Y. Belizario, Jr., Carlos Miguel P. Perez

M alaria remains the leading parasitic disease


that causes mortality worldwide. With
655,000 malaria-related deaths reported in
Table 2.4. Millennium development goals: eight
goals for 2015

Millennium Development Goals


2010 and an estimated 3.3 billion people at
1 Eradicate extreme poverty and hunger
risk for infection, the disease has been identified
2 Achieve universal primary education
by the World Health Organization (WHO)
3 Promote gender equality and women
as one of the three major infectious disease empowerment
threats, along with human immunodeficiency 4 Reduce child mortality
virus/acquired immune deficiency syndrome 5 Improve maternal health
(HIV/AIDS) and tuberculosis, which together
6 Combat HIV/AIDS, malaria, and other diseases
cause more than 5 million deaths each year.
7 Ensure environmental sustainability
Malaria leads to decreased social and economic
8 Develop a global partnership for development
productivity and contributes to a vicious cycle
Source: United Nations. General assembly, 56th session. Road
of disease and poverty. Young children and map towards implementation of the united nations millennium
declaration: report of the secretary-general (UN Document no.
pregnant women are the population groups A1561326). New York: United Nations, 2001.
mostly affected by malaria. Chronic malaria
leads to anemia, which is associated with
impaired physical and mental growth and treated. The group of parasites causing malaria
development in children. In pregnancy, anemia belongs to the genus Plasmodium that is
is a leading contributor to maternal morbidity transmitted by the bite of an infected female
and mortality, and is associated with risk of mosquito belonging to the genus Anopheles.
cardiac failure and adverse perinatal outcomes. The four species that are medically important
Anemia from malaria is also exacerbated by to humans are Plasmodium falciparum, P.
anemia from concomitant helminth infections vivax, P. ovale, and P. malariae. The first two
in both children and pregnant women. are responsible for over 90% of all human
In 2000, the United Nations (UN) adopted malaria cases. More recently, P. knowlesi has
the Millennium Declaration, serving as a been described in humans in the Philippines
blueprint for the eradication of extreme poverty and most of Southeast Asia. P. knowlesi,
through eight quantifiable time-bound targets considered the fifth human malaria parasite,
known as the Millennium Development Goals is normally a parasite of long-tailed macaques
(MDGs) (Table 2.4). MDG 6 aims to reduce (Macaca fascicularis), but humans working in
the burden of HIV/AIDS, malaria, and other nearby forest fringe pose great risk for infection.
diseases. The malaria component of MDG 6 The first naturally acquired human infection
includes reducing incidence and mortality rates was reported in 1965 in Sarawak, Malaysia;
of the disease, increasing insecticide-treated bed other foci of infection have been reported in
net coverage among children below 5 years of Thailand and China as late as 2008. In the
age and increasing anti-malarial coverage among Philippines, the first reported case of P. knowlesi
children below 5 years of age. was described in 2006. Since then, the Research
Despite the high figures in mortality, the Institute for Tropical Medicine (RITM) has
disease is curable if promptly and adequately reported nine cases of mixed malaria infection,
86 MedICal parasItology In the phIlIppInes

positive for P. knowlesi. The life cycle of P. in humans consists of schizogony, which leads to
knowlesi is microscopically indistinguishable the formation of merozoites, and gametogony,
from P. malariae, and differentiation is only which leads to the formation of gametocytes.
achieved through polymerase chain reaction The sexual cycle in the mosquito involves
(PCR) assay and molecular characterization. sporogony, which leads to the formation
These protozoans are pigment producers of sporozoites. The life cycles of all human
and are ameboid in shape, with some being species of malaria are similar. The infected
more ameboid than the others. Their asexual female Anopheles mosquito bites and sucks
cycle occurs in humans, the vertebrate and blood from the human host. In the process,
intermediate host, while the sexual cycle occurs salivary fluids containing sporozoites are also
in the Anopheles mosquito, the invertebrate and injected. These sporozoites, the infective stage
definitive host. of the parasite, are immediately carried to the
liver and enter the parenchymal cells. The
Parasite Biology
parasites then commence exo-erythrocytic
Various processes comprise the life cycle schizogony, which produces the merozoites
(Figure 2.14) of the parasite. The asexual cycle in varying duration and amounts, depending

Figure 2.14. Life cycle of Plasmodium spp.


(Accessed from www.dpd.cdc.gov/dpdx)
Chapter 2: protozoan Infections 87

on the species. Merozoites proceed to the gut as an ookinete, which then develops into
peripheral blood to enter the erythrocytes. Some an oocyst. The oocyst grows and produces
merozoites of P. vivax and P. ovale re-invade the sporozoites, which escape from the oocyst and
liver cells forming hypnozoites, while the other enter the salivary glands of the mosquito. These
species do not. These dormant exo-erythrocytic sporozoites may be injected into another human
forms may remain quiet for years. Within host when the mosquito takes a blood meal.
the red blood cell, the merozoite grows as a The entire developmental cycle in the mosquito
ring form developing into a trophozoite. The takes 8 to 35 days, depending to some extent
trophozoite has an extended cytoplasm and on ambient temperature.
a large chromatin mass which further divides Morphologically, the early trophozoite
to form more merozoites within schizonts. form is ring-shaped with a red chromatin dot
The merozoites of P. falciparum develop in the and a scant amount of blue cytoplasm when
parasitophorous vacuolar membrane (PVM) stained with Giemsa or Wright’s stain. The
within the mature red cells and modify the trophozoite form has a large chromatin mass
structural and antigenic properties of these and a prominent ameboid cytoplasm, which
cells. The parasites feed on the hemoglobin is spread through the erythrocyte. The parasite
resulting in the production of pigment known develops into a schizont when the chromatin has
as hematin. Soon after, the erythrocytes rupture divided into two or more masses of chromatin
and the merozoites are released into the blood, with small amounts of cytoplasm, the so-called
ready to enter new erythrocytes. This asexual merozoites. The number of merozoites is species
cycle is synchronous, periodic, and species- dependent. Clumps of pigment accumulate in
determined. the middle of a mature schizont.
Some merozoites develop into The gametocyte stage fills the entire
microgametocytes (male) or macrogametocytes red blood cell and is characterized by a large
(female) which are picked up by feeding female chromatin mass and a blue cytoplasm with
mosquitoes for completion of the life cycle. In pigment. It is round to banana-shaped. The
the gut of the mosquito, the male gametocytes microgametocyte has a lighter blue cytoplasm,
exflagellate and produce eight sperm-like while the cytoplasm of the macrogametocyte
microgametes which may fertilize the female is a darker blue. Species identification depends
macrogamete to form a zygote. The zygote on various characteristics of these stages of
becomes motile and penetrates the mosquito’s development as described in Table 2.5.

Table 2.5. Comparison of morphological features of malaria parasites

Plasmodium species
Parameter P. falciparum P. vivax P. ovale P. malariae
(malignant tertian) (benign tertian) (benign tertian) (quartan)
Infected red blood Normal: multiple Larger than normal, Somewhat larger than Larger than normal,
cells (RBC) infection of RBC pale, often bizzare; normal, often with pale, often bizzare;
very common Schüffner's dots fringed or irregular Schüffner's dots
are often present; edge, and oval in are often present;
multiple infection shape; Schüffner's multiple infection
of RBC not dots appear even of RBC not
uncommon with younger uncommon
stages; stains more
readily and deeply
than in P. vivax
88 MedICal parasItology In the phIlIppInes

Plasmodium species
Parameter P. falciparum P. vivax P. ovale P. malariae
(malignant tertian) (benign tertian) (benign tertian) (quartan)
Small trophozoite Same as P. vivax Signet-ring form with Small, darker in Same as P. vivax
but with small heavy red dot and color, and but with blue
threadlike blue blue cytoplasmic generally more cytoplasmic circle,
cytoplasmic ring solid than those smaller, thicker and
circle with one of P. falciparum; heavier
or two small red Schüffner's dots
chromatin dots; regularly present
double chromatin in almost 100% of
common; infected cells
marginal forms
common
Growing trophozoite Remains in ring Like small trophozoite, Resembles closely Chromatin rounded
form but grows as above, same stage of or elongated;
resembling small with increased P. malariae but cytoplasm
trophozoite of P. cytoplasm is considerably compact or in
vivax in size; usually and ameboid larger; pigment narrow band
the oldest asexual activity; small- is lighter and less across cell:
stage seen in yellowish brown conspicuous dark brown
peripheral blood pigment granules granules may
in cytoplasm, have peripheral
increasing with arrangement
age of parasite
Large trophozoite Seldom present Large mass of Seldom present Chromatin often
chromatin; elongate,
loose, irregular, or indefinite in outline;
close compact cytoplasm dense,
cytoplasm with compact, in
increasing amount rounded oblong
of fine brown or band forms;
pigment; parasite pigment granules
fills cell in 30 to 40 larger, darker than
hours P. vivax parasite fills
cells frequently
Schizont Not present Chromatin divided; About 25% of Same as P. vivax
(presegmenting) cytoplasm shows infected cells are except parasite
varying degrees definitely oval is smaller, shows
of separation shaped: usual less chromatin
into strands and picture is that of a division, more
particles; pigment round parasite in delayed clumping
collects in parts of the center of an of pigment
the parasite oval cell; many
cells with indefinite
fringed outline;
pigment lighter
and less coarse
than in P. malariae
Schizont Rarely present; 8-24 12-24 merozoites; Usually eight 6-12 (average of
(mature) merozoites; smaller pigment in one merozoites 8-10) merozoites
than other species to two clumps; arranged around in rosette form;
parasite almost fills a central block of parasite almost
enlarged cells pigment fills cell

Pathogenesis and Clinical Manifestations species involved, this may range from 11 days
to 4 weeks. The average pre-patent period for
The interval from sporozoite injection to
P. falciparum is 11 to 14 days, for P. vivax, 11
detection of parasites in the blood is referred
to 15 days, for P. ovale, 14 to 26 days, and 3
to as the pre-patent period. Depending on the
to 4 weeks for P. malariae. The incubation
Chapter 2: protozoan Infections 89

Plasmodium species
Parameter P. falciparum P. vivax P. ovale P. malariae
(malignant tertian) (benign tertian) (benign tertian) (quartan)
Gametocyte Present in peripheral Microgametocyte: Distinguished from P. Same as P. vivax
blood stream, light red to pink malariae by size except smaller; fills
similar to P. vivax; chromatin, of infected cells or almost fills cells
crescent or diffuse, central; and by Schüffner’s
sausage shape gives tint to light dots; less easy to
blue cytoplasm; differentiate from
yellowish P. vivax
brown pigment
throughout
cytoplasm; usually
round and about
the size of normal
RBC
Macrogametocyte:
small, compact,
dark red eccentric
chromatin;
cytoplasm dark
blue, no vacuoles;
abundant dark
brown pigment
scattered
throughout the
cytoplasm
Stages in peripheral Ring forms and All stages present All stages present All stages present
blood gametocytes;
other stages rare
Length of asexual 48 hours or less 48 hours 48 hours 72 hours
cycle
Note: P. knowlesi is microscopically indistinguishable from P. malariae.

period, the time between sporozoite injection with the associated asymptomatic intervals.
and the appearance of clinical symptoms, is Prodromal symptoms may include: a feeling
typically 8 to 40 days, depending again on of weakness and exhaustion, a desire to stretch
the involved species. For P. falciparum, it lasts and yawn, aching bones, limbs, and back, loss
an average of 8 to 15 days, for P. vivax, 12 to of appetite, nausea and vomiting, and a sense of
20 days, for P. ovale, 11 to 16 days, and for P. chilling. At the onset, symptoms may include
malariae, 18 to 40 days. The incubation period malaise, backache, diarrhea, and epigastric
may range from 9 days to 3 years, depending discomfort. The classical malaria paroxysms
on the parasite strain, the dose of sporozoites have three stages: the cold stage, the hot stage,
inoculated, the immune status of the host, and and the sweating stage. The cold stage starts
the host’s malaria chemoprophylaxis history. with a sudden inappropriate feeling of coldness
Partial or incomplete prophylaxis may prolong and apprehension. Mild shivering quickly turns
the incubation period several weeks after to violent teeth chattering and shaking of the
termination of medication. Any person who entire body. Although the core temperature is
has traveled to a malaria-endemic area must be high or may be rising quickly, there is intense
considered at risk of developing malaria up to peripheral vasoconstriction. The patient may
2 years and even longer upon leaving the area. vomit and febrile convulsions may ensue at
There are no absolute clinical features of this stage in young children. These rigors last
malaria except for the regular paroxysms of fever for 15 to 60 minutes after which the shivering
90 MedICal parasItology In the phIlIppInes

ceases, and the hot stage or flush phase begins. distribution. They also depend on the age,
The patient becomes hot and manifests with genetic constitution, state of immunity, general
headache, palpitations, tachypnea, epigastric health and nutritional status of the host, and
discomfort, thirst, nausea, and vomiting. The on any chemoprophylaxis or chemotherapy
temperature may reach a peak of 41°C or even previously used.
more. The patient may become confused or There may be a tendency to recrudesce or
delirious, and the skin may be notably flushed relapse over a period of months to several years.
and hot. This phase lasts from 2 to 6 hours. In Recrudescence is the renewal of parasitemia
the sweating stage, defervescence or diaphoresis or clinical features arising from persistent
ensues with the patient manifesting with undetectable asexual parasitemia in the absence
profuse sweating. The temperature lowers over of an exo-erythrocytic cycle. Relapse is renewed
the next 2 to 4 hours, and symptoms diminish asexual parasitemia following a period in which
accordingly. The total duration of a typical the blood contains no detectable parasites
attack is 8 to 12 hours. The classic periodicity (Figure 2.15). Relapses, which occur with vivax
of attacks develops only if the patient is left and ovale malaria, result from the reactivation
untreated until the time when the life cycle of hypnozoite forms of the parasite in the liver.
phases become synchronized and sufficient Cold, fatigue, trauma, pregnancy, and infections
numbers of red blood cells containing schizonts including intercurrent falciparum malaria may
rupture at about the same time. The interval precipitate reactivation.
between attacks is determined by the length of The pathological processes in malaria
the erythrocytic cycle. For P. falciparum, it is are the result of the erythrocytic cycle. Once
48 hours. For P. vivax and P. ovale, paroxysms the merozoites of P. falciparum invade the
occur on alternate days. For P. malariae, they erythrocytes, the cells reduce their deformability,
occur every 72 hours, causing paroxysms on the degree of which is directly proportional
days 1 and 4, hence the term, quartan malaria. to parasite maturity. This reduction in
Due to the lack of an exoerythrocytic stage in deformability is due to changes in the red
P. knowlesi, fever follows a quotidian pattern, or blood cell cytoskeleton and the increase in
is noted to be non-relapsing. membrane stiffness and cytoplasmic viscosity.
The five species also differ in the age of In the course of invasion, electron-dense sub-
infected erythrocytes. The non-falciparum membranous structures appear and enlarge.
species infect erythrocytes only of a certain These become the so-called “knobs” which are
age: P. vivax and P. ovale infect only young red important in cytoadhesion. They contain several
blood cells, while P. malariae infects only aging proteins such as rosettins, riffins, histidine-
cells. This limits the number of red blood cells rich proteins (HRP), and the Plasmodium
that can be parasitized to less than 3% of all falciparum erythrocyte membrane protein 1
erythrocytes. P. falciparum, as well as P. knowlesi, (PfEMP-1), which is the most adhesive protein
may infect erythrocytes of all ages. As the among the knobs. PfEMP-1 is encoded by a
infected erythrocytes rupture, more falciparum multi-gene family termed var and is clonally
malaria parasites are released to infect more red variant enabling it to evade specific immune
blood cells. The severity of complications and responses. Rosettins and PfEMP-1 are the
mortality increase as the level of parasitemia ligands for rosette formation. They adhere to
increases. The course and severity of the attack parasitized and non-parasitized cells as well
of malaria depend on the species and the strain as blood platelets. In more recent studies, it
of the infecting parasite; therefore, geographical has been suggested that febrile temperatures
origin of infection plays a major role in disease induce the cytoadherence of the ring-staged
Chapter 2: protozoan Infections 91

Figure 2.15. Diagram of the course of malaria infections showing the primary attack, relapses,
and recrudescence (From World Health Organization. Chemotherapy of malaria and resistance to
antimalarials: report of a WHO scientific group. Technical report series no. 529.
Geneva: World Health Organization; 1973.)

P. falciparum erythrocytes, and that the factor cytokines at the time of schizont rupture.
responsible for this heat-induced cytoadherence The combination of altered red cell surface
is PfEMP-1. HRP, on the other hand, localize to membranes and the host’s immunological
the cytoadherence ligands making the adhesion response to the parasite antigens bring about the
more effective. pathologic changes such as alteration in regional
Infected erythrocytes also undergo blood flow in the vascular endothelium, altered
altered membrane transport mechanisms. biochemistry, anemia, and tissue and organ
The hemoglobin is digested forming the hypoxia. Other destructive tissue processes
pigment hematin, and variant strain-specific include increased capillary permeability which
neoantigens are expressed. The soluble antigens allows fluid to leak into surrounding tissues, and
of P. falciparum are potent inducers of pro- congestion in blood vessels resulting in tissue
inflammatory as well as anti-inflammatory infarction and necrosis.
cytokines from monocytes and macrophages. In severe forms of malaria, impairment
Glycosylphosphatidyl inositol (GPI) moieties of consciousness and other signs of cerebral
that are seen covalently linked to the surface dysfunction, such as delirium and generalized
antigens of these protozoans act like the convulsions, are commonly observed. Other
endotoxin of gram-negative bacteria, manifestations are severe hemolytic anemia
lipopolysaccharide (LPS). They stimulate the with a hematocrit less than 20%, hemoglobin
monocytes to release tumor necrosis factor levels less than 7 g/dL and hyperbilirubinemia
(TNF) or cachexin, which is implicated as with levels more than 50 mmol/L (Table 2.6).
the cause of malarial fever. The fever, febrile Cerebral malaria generally manifests with
paroxysms, headache, various aches and pains, diffuse symmetric encephalopathy. Other signs
and prostration, which are the more familiar and symptoms include retinal hemorrhage,
symptoms of an acute malarial attack, are bruxism (fixed jaw closure and teeth grinding),
probably the result of the release of these and mild neck stiffness. Pouting may occur or
92 MedICal parasItology In the phIlIppInes

Table 2.6. Clinical features and laboratory findings in severe malaria infection

Clinical features Laboratory results


• Impaired consciousness or coma • Hypoglycemia (blood glucose <2.2 mmol/L or <40 mg/
• Prostration dL)
• Failure to feed • Metabolic acidosis (plasma bicarbonate <15 mmol/L)
• Multiple convulsions* • Severe normocytic anemia (Hb <5 g/dL, packed cell
• Deep breathing volume <15%)
• Respiratory distress • Hemoglobinuria
• Circulatory collapse or shock (systolic blood pressure • Hyperparasitemia (>2% or 100 000/μL in low intensity
below 50 mmHg in children) transmission areas or >5% or 250,000/μL in areas of high
• Clinical jaundice stable malaria transmission intensity)
• Other evidence of vital organ dysfunction • Hyperlactatemia (lactate >5 mmol/L)
• Abnormal spontaneous bleeding • Renal impairment (serum creatinine >265 μmol/L)
• Pulmonary edema (radiological)
*more than two episodes in 24 hours Note: Severe P. falciparum infection, one or more clinical feature and
laboratory finding

Source: World Health Organization. Management of severe malaria: a practical handbook. Geneva: World Health Organization; 2000.

a pout reflex may be elicited by stroking the affected. Malaria ARF is defined as having a
sides of the mouth. Lumbar tap usually reveals serum creatinine of more than 265 mmol/L
a normal to elevated opening pressure, clear (3 mg/dL) and a 24-hour urine output of
cerebrospinal fluid (CSF) with fewer than 10 less than 1 ml/kg/hr, despite rehydration, in
leukocytes/mL, and slightly elevated protein and patients with asexual forms of the parasite
CSF lactic acid concentration. If left untreated, present in their peripheral blood smear. The
symptoms progress to persistent coma and patient may also present with hyperkalemia
death. The neurological complications, once and hyperuricemia earlier in the course.
promptly and adequately treated, are reversible The cytoadherence, rosette formation, and
and a majority of the patients make a complete sequestration of parasitized erythrocytes lead
recovery. to a decrease in tissue perfusion resulting in
Respiratory findings are also a major decreased renal blood flow. The increase of TNF
feature of severe malaria. Altered pulmonary in tubular epithelial cells leads to inflammatory
function is common, and it includes air flow cell infiltration in the interstitium and altered
obstruction, impaired ventilation and gas tubular transport, which result in tubular
transfer, and increased pulmonary phagocytic damage and dysfunction. The presence of GPI
activity. In African children, pneumonitis from and other falciparum malaria antigens lead to
sequestered, parasitized RBC and inflammatory release of cytokines and mediators that decrease
cells are seen in postmortem pulmonary the systemic vascular resistance and increase
vasculature, while in adults, non-cardiogenic renal vascular resistance. All these changes
pulmonary edema and acute pulmonary distress eventually lead to acute tubular necrosis causing
syndrome (ARDS) may be observed. There is a acute renal failure.
high mortality rate (over 80%) when pulmonary Malaria in pregnancy increases the risk of
edema develops in a patient with severe malaria. maternal death, maternal anemia, intrauterine
Factors which predispose to pulmonary edema growth retardation, spontaneous abortion,
include hyperparasitemia, renal failure, and stillbirth, and low birth weight associated with
pregnancy. risk for neonatal death. Non-immune pregnant
The incidence of acute renal failure (ARF) women are susceptible to all complications
reaches up to 60% of patients with severe associated with severe malaria such as cerebral
falciparum malaria, with more males being malaria, hypoglycemia, and pulmonary edema.
Chapter 2: protozoan Infections 93

For partially immune pregnant women, Not everyone infected with the malaria
especially primigravid, severe anemia may parasite becomes seriously ill or dies. In areas
develop but the other complications of severe where endemicity is stable, repeated exposures
malaria are unlikely to occur. Falciparum to the parasite lead to specific immunity.
malaria may induce uterine contractions, thus This restricts occurrence of serious problems
may push the patient to premature labor. In in young children, while older patients have
severe malaria, the prognosis of the fetus is poor. relatively mild febrile illness. In people who are
Falciparum malaria in a young child is exposed to malaria for the first time, possible
considered a medical emergency for it can be outcomes may range from apparent resistance to
rapidly fatal. The initial symptoms may be death. Any resistance, therefore, is nonspecific.
atypical and difficult to recognize, but within It also does not necessarily depend on prior
hours, life-threatening complications may start exposure to malaria and may be either acquired
to occur. The most common complications or innate. Poor prognostic factors in falciparum
of severe malaria in children are cerebral malaria include hyperparasitemia defined as
malaria, severe anemia, respiratory distress, and a peripheral count more than 250,000/µL or
hypoglycemia. Children with severe malaria more than 5% of the RBCs infected, and the
most commonly present with seizures. These presence of mature or immature schizonts in
convulsions are common before or after the a peripheral blood smear. It has been shown
onset of coma and are significantly associated that a peripheral count of 10% or more of
with neurologic sequelae. Opisthotonos may red blood cells infected has a mortality rate
also be observed in some children. As much as of 50%, particularly in non-immune cases,
10% of children who survive cerebral malaria despite treatment. The clinical indicators of
will develop sequelae such as hemiparesis, poor prognosis include deep coma, absence
cerebellar ataxia, speech disorders, generalized of corneal light reflex, respiratory distress
spasticity, or some behavioral disturbances (acidosis), circulatory collapse, decerebrate
(Table 2.7). or decorticate rigidity, opisthotonos, and age

Table 2.7. Comparison of sign and symptoms of sever malaria in adults and children

Sign or symptom Adults Children


History of cough Uncommon Common
Convulsions Common Uncommon
Duration of illness 5-7 days 1-2 days
Resolution of coma 2-4 days 1-2 days
Neurological sequelae <5% >10%
Jaundice Common Uncommon
Pretreatment hypoglycemia Uncommon Common
Pulmonary edema Uncommon Common
Renal failure Common Uncommon
CSF opening pressure Usually normal Usually raised
Respiratory distress (acidosis) Sometime Common
Bleeding/clotting disturbances Up to 10% Rare
Abnormal brainstem reflex (e.g., oculovestibular, oculocervical) Rare More common
Source: World Health Organization. Management of severe malaria: a practical handbook. Geneva: World Health Organization; 2000.
94 MedICal parasItology In the phIlIppInes

below 3 years. Other laboratory indicators of


poor prognosis include blood glucose <2.2
mmol/L, raised venous lactic acid (>5 mmol/L),
more than three-fold increase in serum enzymes
(aminotransferases), hemoglobin concentration
less than 5g/dL, blood urea more than 60 mg/
dl, serum creatinine more than 265 mmol/L,
peripheral polymorphonuclear leukocytes
with visible malaria pigment (>5%), low
antithrombin III levels, and very high plasma
concentrations of TNF.
Diagnosis Plate 2.16. Plasmodium falciparum ring forms
(Courtesy of the Department of Parasitolgy,
Prompt and adequate diagnosis of malaria UP-CPH)
is necessary for the disease to be managed
effectively, thus preventing the life threatening are not seriously ill, monitoring once daily
complications. Though malaria may present may be sufficient. Seriously ill patients should
with the classic paroxysms of fever with be monitored two to three times daily until
asymptomatic intervals, initial symptoms are significant improvement occurs. Monitoring
non-specific and are not reliable in clinching the should be continued until there is clearance of
diagnosis. In fact, treatment based on clinical parasitemia.
findings alone usually results in unnecessary and Although microscopic diagnosis is the
irrational drug use. established diagnostic method, technical and
Microscopic identification of the malarial personnel requirements often cannot be met,
parasites in thick and thin blood smears stained particularly facilities in the periphery of the health
with Giemsa or Wright’s stain is still important care system. This has led to the introduction of
in making the definitive diagnosis and remains the malaria rapid diagnostic tests (RDTs). These
as the gold standard. Specimens may be taken tests make use of immunochromatographic
any time and all blood stages of the parasite may methods in order to detect Plasmodium-
be found. In falciparum malaria, only the ring specific antigens in a finger prick blood sample.
forms (Plate 2.16) may be found, but 10 days Currently, the antigens being targeted by these
after the symptoms begin, gametocytes may be RDTs include: histidine rich protein II (HRP
found as well. Although there are no standard II), which is a water soluble protein produced
recommendations on how often the blood by trophozoites and young gametocytes of P.
smears should be taken in order to diagnose falciparum; Plasmodium lactate dehydrogenase
malaria, obtaining smears every 6 to 8 hours (pLDH), which is produced by both sexual
is usually appropriate. This may have to be and asexual stages and can distinguish between
continued until a diagnosis of malaria is made P. falciparum and non-P. falciparum, but not
or until malaria can be confidently ruled out. among the non-P. falciparum species; and
When malaria is a serious condition, this may Plasmodium aldolase, an enzyme in the parasite
require repeated testing for several days in order glycolytic pathway expressed by the blood
to demonstrate a positive result. Even after the stages of all Plasmodium species. Together
diagnosis of malaria has been made, peripheral with HRP II, Plasmodium aldolase has been
blood smears should still be obtained to monitor used in a combined immunochromatographic
the response to treatment. In individuals who test targeting the panmalarial antigen (PMA).
Chapter 2: protozoan Infections 95

These tests can be performed in 15 to 30 differentiate between current and past infections
minutes without the use of electricity, special and are therefore most helpful in epidemiologic
equipment, or any training in microscopy, and studies. Current studies are using PCR to
most kits have more than 90% specificity. More significantly enhance microscopic diagnosis of
recent studies have shown that test kits based malaria especially in cases of low parasitemia
on HRP II have a sensitivity and specificity of and in cases of mixed infection.
92.5% and 98.3% respectively, while kits based
Treatment
on the pLDH antigen have a lower sensitivity
(88.5%) albeit a higher specificity at 99.4%. Antimalarial drugs have selected actions
The use of RDTs can be easily taught to village on the different phases of the life cycle of the
health workers and the results can likewise be malaria parasite. These drugs may be classified
easily interpreted. The main disadvantages into causal prophylactic drugs, which prevent
of RDTs compared to microscopy are: the the establishment of the parasite in the liver,
lack of sensitivity at low levels of parasitemia; and blood schizonticidal drugs, which attack
the inability to quantify parasite density; the the parasite in the red blood cell, preventing
inability to distinguish among P. vivax, P. ovale, or terminating the clinical attack. Tissue
and P. malariae, as well as sexual and asexual schizonticides act on pre-erythrocytic forrns
stages; the persistently positive tests (for some in the liver. Gametocytocidal drugs destroy
antigens) despite parasite clearance following the sexual forms of the parasite in the blood.
chemotherapy; and the relatively higher cost Some drugs are hypnozoitocidal or antirelapse
per test. drugs, which kill the dormant forms in the liver.
In recent studies conducted in various areas Sporonticidal drugs inhibit the development
of the Philippines to describe the validity of a of the oocysts on the gut wall of the mosquito,
few specific malaria RDT kits, results showed which has fed on a gametocyte carrier so that
sensitivity and specificity levels below the the mosquito cannot transmit the infection.
WHO recommended ideal of 95% and 90%, The main uses of antimalarial drugs are:
respectively. Reasons for these findings could be (a) protective (prophylactic), (b) curative
manufacturer-related problems, the instability (therapeutic), and (c) preventive. Drugs for
of the substances used in the diagnostic prophylaxis are used before the infection occurs
technique to varying environmental conditions or before it becomes evident, with the aim of
such as extremes of temperature and humidity, preventing either the occurrence of the infection
and user-related problems. Quality assurance of or any of its symptoms. A blood schizonticidal
these malaria RDT kits is, therefore, necessary drug may have minimal effects on parasites
before they are deployed on a larger scale in growing in the liver, but if it is still present in
remote and rural areas. More recent studies are the blood when the merozoites leave the liver
now concentrating on quality assurance of these and invade the blood cells for the first time, it
tests and on identifying the factors which may will effectively prevent symptomatic malaria.
affect RDT performance in the field. Curative or therapeutic use refers to action on
Malaria can also be diagnosed serologically the established infection, which involves the use
but presently available methods are not of blood schizonticidal drugs for the treatment
capable of making a definitive diagnosis of of the acute attack and in the case of relapsing
acute malaria. Available serologic tests like malaria, radical treatment of the dormant liver
indirect hemagglutination (IHA), indirect forms. Prevention of transmission means the
fluorescent antibody test (IFAT), and enzyme- deterrence of infection of mosquitoes with
linked immunosorbent assay (ELISA) cannot the use of gametocytocidal drugs to attack the
96 MedICal parasItology In the phIlIppInes

gametocytes in the blood of the human host. It be given in pregnancy and in children less than
also means the interruption of the development 4 years of age.
of the sporogonic phase in the mosquito when In contrast with falciparum malaria, vivax
it feeds on the blood of an infected person who malaria remains sensitive to chloroquine. Clinical
has been given the appropriate sporonticidal studies and extensive in vitro observations have
compound. shown that P. vivax is still generally sensitive to
Chloroquine was the mainstay of chloroquine, although resistance is prevalent
antimalarial treatment for the last 50 years. and increasing in Indonesia, Peru, and Oceania.
Because of emergence of multidrug-resistant Moreover, vivax malaria is sensitive to all other
(MDR) strains, subsequent chloroquine use antimalarial drugs albeit slightly less sensitive to
has been rendered ineffective against falciparum artesunate plus sulfadoxine-pyrimethamine. The
malaria, and the current DOH Malaria asexual stage of P. vivax remains susceptible to
Control Program (MCP) recommends the use primaquine; therefore, combination treatment
of artemisinin-based combination therapies with chloroquine and primaquine affords blood
(ACTs) for severe and uncomplicated falciparum stage and liver stage treatment, respectively.
malaria, replacing the chloroquine plus Often referred to as radical treatment, the use of
sulfadoxine-pyrimethamine combination. The primaquine, together with chloroquine, allows
following drug combinations are recommended: for the prevention of relapse in vivax malaria. In
artemether plus lumefantrine, artesunate plus comparison with no primaquine treatment, the
amodiaquine, artesunate plus mefloquine, risk of relapse decreases for every additional mg/
and artesunate together with sulfadoxine- kg of primaquine given. Repeated vivax malaria
pyrimethamine. For severe malaria, parenteral relapses are debilitating at any age, hence they
antimalarial treatment should be started must be prevented. At least a 14-day course of
without delay after rapid clinical assessment and primaquine is needed for the radical treatment
confirmation of the diagnosis. The following of P. vivax.
antimalarial drugs are recommended: artesunate Resistance of P. malariae and P. ovale
intravenous (IV) injection or intramuscular to antimalarials is not well characterized,
(IM) injection, quinine IV or IM, or artemether and infections with these species are still
IM. In a placebo-controlled trial, patients with considered sensitive to chloroquine. The
severe malaria who could not be treated orally treatment for relapsing fever caused by P.
and where access to IM and IV treatment was ovale is similar to that of vivax malaria (i.e.,
unavailable, a single artesunate suppository at chloroquine and primaquine). In the case of
the time of referral reduced the risk of death or mixed malarial infections, ACTs remain the
permanent disability. mainstay treatment. Moreover, the use of
Current guidelines also recommend artemisinin-based compounds and a partner
the use of gametocytocidal drugs to reduce drug with a long half-life (i.e., artesunate
transmission. Seen in the context of malaria plus amodiaquine and dihydroartemisinin
elimination, the use of primaquine 0.75 plus piperaquine) has been effective against
mg base/ kg body weight single oral dose in chloroquine-resitant vivax malaria. Radical
demonstrates an added benefit to artemisinins treatment with primaquine should also be
in eliminating gametocytes. The addition of a considered in cases of confirmed P. vivax and
single dose of primaquine to current ACTs is P. ovale infections.
therefore recommended provided that the risk Artemisinin and its derivatives (Qinghaosu
for hemolysis in G6PD deficient patients is derivatives), artesunate, and artemether produce
considered. Moreover, primaquine should not rapid clearance of parasitemia and rapid
Chapter 2: protozoan Infections 97

resolution of symptoms. Because artemisinins of therapy. Resistance of a parasite to drugs


are rapidly eliminated in the body, the duration is graded according to the patterns of asexual
of treatment is dependent on the partner parasitemia after initiation of treatment (Figure
drug being short acting or long acting. When 2.16). RI is the mildest form of resistance which
partnered with rapidly eliminated drugs is characterized by initial clearance of parasites
(tetracyclines and clindamycin), a 7-day course but recrudescence occurs within a month after
of treatment is usually required. Treatment the start of treatment. It can be classified as
duration can be reduced to 3 days when either early, when clearance occurs for the first
artemisinins are given in combination with 48 hours and recrudescence takes place within
slowly eliminated drugs such as mefloquine the first 14 days after start of treatment, or late
and amodiaquine. An additional advantage when there is also clearance within the first 48
from a public health standpoint is the ability hours and recrudescence occurs within the 14th
of artemisinins to reduce gametocyte carriage, to the 28th day from the start of treatment. RII
thus reducing the transmission of malaria. This
form of malaria control is particularly useful in
areas of low to moderate endemicity.
Quinine sulfate plus doxycycline or
clindamycin serves as the second line drug when
artemisinins (e.g., IV artesunate) are unavailable
or when there is failure to respond to artemisinin
therapy. The tetracyclines and clindamycin are
known to be effective antimalarials, although
they kill the parasite rather slowly. Quinine
has the disadvantage of producing toxic side
effects such as cardiotoxicity and cinchonism,
characterized by tinnitus, headache, and
blurring of vision. Also, rapid administration
of quinine is unsafe. Each dose of parenteral
quinine must be administered as a slow rate-
controlled infusion, and electrocardiographic
(ECG) monitoring and frequent assessment of
vital signs are required if quinines are used. Due
to the risk of congenital defects, tetracycline
is contraindicated in pregnant women and
children below 8 years. Rather, quinine plus
clindamycin taken for 7 days remains the
antimalarial of choice in pregnancy.
The problem of drug resistance involves
mainly chloroquine plus sulfadoxine- Figure 2.16. A WHO field test for response of
pyrimethamine and certain strains of P. malaria parasites to chloroquine
falciparum. Such strains are often MDR. (From World Health Organization. Chemotherapy
of malaria and resistance to antimalarials:
Asexual parasites are normally cleared from report of a WHO scientific group. Technical
the blood three days after the start of treatment report series no. 329. Geneva: World Health
and are definitely cleared 6 or 7 days after start Organization; 1973.)
98 MedICal parasItology In the phIlIppInes

shows an initial reduction in parasitemia after without previously meeting any of the criteria
treatment but there is failure to clear the blood of ETF; and (b) presence of parasitemia and
of asexual parasites and soon after an increase an axillary temperature of 37.5°C (or history
of parasitemia follows. RIII is the severest form of fever) on any day from Day 4 to Day 28,
of resistance in which parasitemia will either without previously meeting any of the criteria
show no significant change with treatment or for ETF. Late parasitological failure for intense
will eventually increase. transmission areas is defined as presence of
MDR malaria is considered when treatment parasitemia on Day 14 and axillary temperature
failure occurs with three or rnore antimalarial of 37.5°C without previously meeting any of the
agents. In this case, a combination of artesunate criteria for ETF or late clinical failure.
has been combined with mefloquine and is now For low to moderate transmission areas,
the first-line regimen for MDR malaria in some late parasitological failure is defined as presence
Southeast Asian countries. of parasitemia on any day from Day 7 to Day
Classification of response to malaria 28 and axillary temperature of 37.5°C without
treatment can be divided into early treatment previously meeting any of the criteria for ETF
failure, late treatment failure, and adequate or late clinical feature. Adequate clinical and
clinical and parasitological response. Early parasitologic response for intense transmission
treatment failure (ETF) is present when there areas is defined as absence of parasitemia on Day
is: (a) development of danger signs or severe 14 (Day 28 for low to moderate transmission
malaria on Day 1, 2, or 3 in the presence of areas) irrespective of axillary temperature
parasitemia; (b) parasitemia on Day 2 higher without previously meeting any of the criteria of
than the Day 0 count irrespective of axillary ETF, late clinical failure, or late parasitological
temperature; (c) parasitemia on Day 3 with failure.
axillary ternperature of 37.5°C; and (d) In cases of renal failure in severe malaria,
parasitemia on Day 3 which is 25% of count dopamine may be given at 3 to 5 µg/kg/
on Day 0. This classification of ETF holds for minute. If the patient remains unresponsive
both intense transmission and low to moderate despite adequate rehydration and other forms
transmission areas. Late treatment failure (LTF) of therapeutic management, and blood urea
is further divided into late clinical failure and and creatinine are rising progressively, dialysis
late parasitological failure. The definitions for is indicated.
these two would differ depending on whether For control of seizures, diazepam may be
the area is an intense transmission area or a given at 10 mg intravenous (pediatric dose at
low to moderate one. Late clinical failure for 0.3 mg/kg IV up to 10 mg) or in cases of status
intense transmission areas is defined as: (a) epilepticus, phenytoin at a loading dose of 13
development of danger signs or severe malaria to 18 mg/kg and a maintenance dose of 3 to 5
after Day 3 in the presence of parasitemia, mg/kg per day (pediatric dose: loading dose of
without previously meeting any of the criteria 15-20 mg/kg slow IV push and maintenance
of ETF; and (b) presence of parasitemia and dose of 5 mg/kg in two divided doses). The
axillary temperature equal to or greater than following are now not considered useful and are
37.5°C on any day from Day 4 to Day 14, therefore not recommended in the management
without previously meeting any of the criteria of cerebral malaria: corticosteroids, other anti-
for ETF. For low to moderate transmission inflammatory agents, low molecular weight
areas, late clinical failure is defined as: (a) dextran, epinephrine, and heparin.
development of danger signs or severe malaria Proper management of malaria also
after Day 3 in the presence of parasitemia, includes general and supportive measures
Chapter 2: protozoan Infections 99

especially in P. falciparum infections. If fluid malaria have shrunk considerably over the past
replacement or blood transfusion is necessary, 50 years, but control is becoming more difficult,
it must be administered with care to avoid and past gains have been threatened. The spread
pulmonary edema. Antipyretics and sponging of the disease is linked to activities like road
for high fever are important especially in building, mining, logging, and new agricultural
children to prevent convulsions. Blood sugar and irrigation projects, particularly in “frontier”
should be monitored regularly especially in areas (e.g., forest fringe, mountain valleys and
severe malaria. If hypoglycemia develops, 50 reclaimed areas). Elsewhere, disintegration
mL of 50% dextrose (1.0 mL/kg for children) of health services, armed conflict, and mass
diluted in an equal volume of infusion fluid movements of refugees have worsened the
should be infused over a 5-minute period, malaria situation.
followed by a continuous intravenous infusion Malaria remains a public health problem
of 5 to 10% dextrose. today in more than 90 countries inhabited by
a total of some 3.3 billion people (Figures 2.17
Epidemiology
to 2.19). Of these, 2.1 billion are at low risk
Malaria is the world’s most important (<1 case per 1,000 population), 94% of whom
tropical parasitic disease. The disease kills live in areas outside the WHO African region.
more people than any other communicable The 1.2 billion at high risk (>1 case per 1,000
disease except tuberculosis. In many developing population) live in the WHO African (47%)
countries, especially in Africa, malaria has an and Southeast Asian Regions (37%).
enormous toll on lives, medical costs, and days In 2010, the WHO reported an estimated
of labor lost. The geographical areas affected by 216 million cases of malaria, of which 81% or

Figure 2.17. Global distribution of malaria: malaria-free and malaria-endemic countries in phases of
control, elimination and prevention of reintroduction (From World Health Organization. World Malaria
Report 2009. Geneva: World Health Organization; 2009.)
100 MedICal parasItology In the phIlIppInes

Figure 2.18. Distribution of malaria in the WHO Southeast Asia Region: areas in red indicate malaria-
endemic countries (Accessed from http://www.map.ox.ac.uk)

deaths occurred in 2010, 91% of which were


in Africa, and approximately 86% of these
deaths were children under 5 years of age.
The estimated incidence of malaria has fallen
by 17% globally between 2000 and 2010.
Large percentage reductions were seen in the
European (99.5%), American (60%), and
Western Pacific (86%) WHO regions. Likewise,
malaria specific mortality rates have fallen by
25% between 2000 and 2010.
According to the World Malaria Report
2011, the WHO cites a decreasing number
of malaria cases in a majority of countries
belonging to the Western Pacific Region. A
greater than 50% decrease in cases were reported
for China, Philippines, Republic of Korea,
Figure 2.19. Distribution of malaria in the WHO Solomon Islands, and Vietnam, while a 25-50%
Western Pacific Region: areas in red indicate decrease in the number of cases were seen in Lao
malaria-endemic countries People’s Democratic Republic, Malaysia, and
(Accessed from http://www.map.ox.ac.uk)
Vanuatu. No notable change in the number of
malaria cases were seen in Cambodia and Papua
171 million cases where in the African region, New Guinea.
with the Southeast Asian Region accounting for In the Philippines, malaria has not been
another 13%. An estimated 655,000 malaria included among the 10 leading causes of
Chapter 2: protozoan Infections 101

Figure 2.20. Malaria cases per 100,000 population in the Philippines from 2000 to 2009
(From Department of Health-National Center of Disease Prevention and Control. Malaria medium term
development plan 2011-2016. Manila (Philippines): Department of Health; 2011.)

morbidity since 2006. Cases have been notably reported in 2009 (Figure 2.21). Similarly, the
decreasing as reported in 2009 (Figure 2.20). malaria morbidity rate has decreased by 58.3%,
However, disease prevalence, seen in the 2010 with 18,781 malaria cases reported in 2009.
DOH-Malaria Control Program (MCP) Among blood smears examined from 2005 to
report, remains markedly high in Regions 2009, 69.4 to 80% of patients were diagnosed
IV-B, Caraga, III, XII, and II. There remains with P. falciparum, 17.0 to 23.4% with P. vivax,
an estimated 10.8 million people still at risk approximately 1% with P. malariae, and a small
for the disease, consisting mostly of farmers, number of cases (0.5%) were diagnosed to have
indigenous cultural groups, miners, forest mixed malaria infection, falciparum and vivax
product gatherers, and soldiers. Fifty nine out of malaria being the usual mixed infection.
the 80 provinces in country are endemic for the Macrostratification of malaria endemic
disease, with 60.4% of the endemic provinces areas serves to classify the different provinces
located in Luzon, 39.5% in Mindanao, and based on annual incidence of the disease
0.1% in Visayas. As of 2009, the provinces of in each respective province (Table 2.8).
Cagayan, Isabela, Palawan, Sulu, and Tawi-Tawi Macrostratification provides an opportunity
comprise the five provinces having the highest for planning, policy making, and resource
number of malaria cases reported. allocation of the provincial MCP. The number
It appears that in areas of low malaria of provinces in Category A has been reduced
endemicity, there is a clustering of cases, from 26 in 2000, to nine in 2005 and finally to
resulting in pockets of high endemicity. five in 2008. The values reported for Category
Mortality rate for malaria has markedly B provinces have changed from 22 in 2000 to
decreased by 88.2% from 2005 figures to values 31 in 2005, and to 27 in 2008. Malaria-free
102 MedICal parasItology In the phIlIppInes

Figure 2.21. Malaria-related deaths per 100,000 population in the Philippines from 2005 to 2009
(From Department of Health-National Center of Disease Prevention and Control. Malaria medium term
development plan 2011-2016. Manila (Philippines): Department of Health; 2011.)

Table 2.8. Macrostratification of malaria Philippines, the principal malaria vector is


endemic provinces according to annual Anopheles minimus var. flavirostris, a night biter,
incidence of malaria
which prefers to breed in slow flowing, partly
Category Annual incidence of malaria shaded streams that abound in the foothill areas.
A ≥1000 cases
Occasionally, it has the ability to adapt to or
B 100 to <1000 cases
utilize new habitats such as irrigation ditches,
C <100 cases
rice fields, pools, and wells. In Palawan, it was
observed to be mildly exophagic and zoophilic.
D No documented indigenous case for the
past 5 years Its horizontal flight range has been reported
Source: Department of Health. Administrative Order no. 14 series to be about 1 to 2 km. Anopheles litoralis is
of 1996: Technical guidelines on stratification of areas. 1996.
associated with malaria transmission in the
coastal areas of Mindanao, particularly in Sulu.
provinces have increased from 13 in 2000, to Anopheles maculatus coexists with A. flavirostris
22 in 2009. Four provinces in Category A, eight in the portion of streams exposed to sunlight.
provinces in Category B and eight provinces They appear to be responsible for malaria
in Category C have been reclassified to the transmission at higher altitudes. Anopheles
immediate lower categories respectively, from mangyanus has the same breeding habitats and
2005 to 2009. Nueva Ecija is noted to have seasonal prevalence as A. flavirostris but appears
shifted into a higher category (Figure 2.22). to prefer habitats located in forest fringe.
Peak transmission occurs at the beginning Malaria can also be transmitted through
and at the end of the rainy season. In the blood transfusion from infected donors, and
Chapter 2: protozoan Infections 103

Figure 2.22. Macrostratification of provinces in the Philippines according to category by average


malaria cases: note that the Isabela, Cagayan, Palawan, Sulu, and Tawi-Tawi remain
Category A provinces as of 2008 (From Department of Health-National Center for Disease Prevention
and Control. Malaria control program. 2009.)

by contaminated needles and syringes. Blood breeding sites, and estimation of mosquito
from semi-immune donors without clinical density.
symptoms may contain malaria parasites. In
Prevention and Control
congenital malaria, infected mothers transmit
parasites to their child before or during birth. Early diagnosis and prompt treatment
The evaluation of the amount and of malaria are essential for malaria control.
conditions of transmission of malaria in a Breeding sites of Anopheles flavirostris should
given locality is called the malaria survey. be detected early and contained. Personal
Disease control efforts must always take into protection measures against mosquito bites
consideration the findings of the malaria are also helpful and cost-effective. The use
survey. The survey involves looking into of insecticide- (permethrin or deltamethrin)
epidemiologic data regarding the disease, such as treated nets (ITNs) and long lasting insecticide-
malaria mortality and morbidity, investigations treated nets (LLIN) remains the major vector
relating to the human host, and investigations control strategy coupled with indoor residual
relating to the insect vector. Investigations spraying (IRS), with the latter used in epidemic
relating to the human host include blood and situations, areas with stable transmission but
spleen examinations. Investigations relating without reduction of malaria incidence, and
to the vector, on the other hand, may include areas of displaced populations. Wearing of
identification of mosquito vectors and their light colored clothing, which cover most of the
104 MedICal parasItology In the phIlIppInes

Table 2.9. Treatment of malaria infection

Common name Chemical class Clinical use Resistance


Artemisinins Sesquieterpine lactone In artemisinin-based Possibly emerging
(artemether, artesunate, endoperoxide combination therapies
dihydroartemisinin) (ACTs)
Lumefantrine Arylamino alcohol Most common first line No evidence of high level
anti-malarial therapy in resistance
Africa, in combination
with artemether
Artemether plus
lumefantrine (AL) –
most common drug
combination used
in uncomplicated
falciparum malaria
Amodiaquine 4-aminoquinoline In combination with Limited cross resistance with
artesunate in parts of chloroquine
Africa
Piperaquine Bisquinoline In combination with Observed in China following
dihydroartemisinin in single drug therapy
parts of Southeast Asia
Mefloquine 4-methanolquinoline In combination with Prevalent in Southeast Asia
artesunate in parts of
Southeast Asia
Pyronaridine Acridine type Mannich base Being registered for No cross-resistance with
combined use with other drugs
artesunate
Quinine/quinidine 4-methanolquinoline Mainly used for the Exists at a low level
treatment of severe
malaria, often in
combination with other
antibiotics
Drug of choice in severe
malaria
Atovaquone Naphthoquinone In combination with Has been observed
proguanil (a biguanide) clinically
for treatment or
prevention
Chloroquine 4-aminoquinoline Former first line treatment, Widespread
together with sulfadoxine-
pyrimethamine (SP)
of uncomplicated
falciparum malaria
Remains drug of choice for
vivax malaria
Pyrimethamine Diaminopyrimidine For intermittent preventive Widespread
treatment, combined
with sulfadoxine (a
sulfonamide)
Primaquine 8-aminoquinoline For eliminating liver-stage Unknown
parasites, including
dormant forms of P. vivax
Drug of choice for
gametocytes and
hypnozoites
Source: Fidock DA. Drug discovery: priming the antimalarial pipeline. Nature. 2010;465: 297-298.
Chapter 2: protozoan Infections 105

body (since dark colors attract mosquitoes), and rice fields and bacterial insecticide (PG-14
using insect repellants containing DEET Bacillus thuringiensis), and chemical control
(N,N-diethyl-m-toluamide, optimally as a 35% such as the use of mosquito repellants and
concentration lotion) on exposed parts of the insecticide treated mosquito nets.
body, using a insect spray containing pyrethrum In the field of molecular entomology,
in living areas, and use of permethrin insecticide stable germline transformation of the Anopheles
as a repellant spray for clothing have known to mosquito is being investigated. This research
be effective personal protection measures as well. involves inserting genes (e.g., immune response
Chemoprophylaxis may be protective to genes) that will inhibit the development of
travelers who have no immunity to malaria, the parasite in the mosquito. With the recent
although no chemoprophylactic regimen sequencing of the genomes of Plasmodium
ensures 100% protection. Because of this, falciparum and of the Anopheles mosquito,
precautions to avoid mosquito bites are new areas of research for malaria treatment and
needed even if antimalarials have been taken. prevention are now being explored.
Prophylactic drugs should be taken with good
References
compliance for the duration of the stay and
should be continued four weeks after the last Anstey NM, Jacups SP, Cain T, Pearson, T,
possible exposure to infection since the parasites Ziesing PJ, Fisher, DA, et al. Pulmonary
may still emerge from the liver after this period. manifestations of uncomplicated
An exception would be atovaquone/proguanil falciparum and vivax malaria: cough, small
which can be stopped one week after return. airways obstruction” impaired gas transfer,
Chloroquine is only recommended for areas and increased pulmonary phagocytic
where malaria is exclusively due to P. vivax or activity. J Infect Dis. 2002;85(9):1326–34.
where there is low risk of chloroquine-resistant Beeston JG, Brown GV. Pathogenesis of
P. falciparum. Those traveling to areas where Plasmodium falciparum malaria: The roles
levels of resistance to chloroquine are high may of parasite adhesion and antigenic variation.
use mefloquine, doxycycline or atovaquone/ Cell Mol Life Sci. 2002;59(2):258–27.
proguanil. Strategies implemented to control Belizario VY, Pasay CJ, Bersabe MJ, de Leon
malaria in pregnancy include the use of ITNs WU, Guerrero D, Alcantara S, et al.
and intermittent preventive treatment, which Field and laboratory evaluation of rapid
involves providing all pregnant women with at diagnostic test for malaria. Final report
least two preventive treatments of an effective 2003. Geneva: World Health Organization;
antimalarial drug. 2003.
Work is on-going for the development of Belizario VY, Saul A, Bustos MDG, Lansang,
an effective malaria vaccine. Among the vaccine MA, Pasay, CJ, Gatton M, et al. Field
types being developed are the “sporozoite” epidemiologic studies on malaria in a
vaccines, “asexual” vaccines, and the “altruistic” or known endemic community in Bataan.
“transmission blocking” vaccines. Combination Acta Trop. 1997;63:257–66.
vaccines derived from multiple parasite life stages Bustos, MD. Clinical aspects, chemotherapy,
are also being developed. and drug resistance. In: Philippine Council
Malaria control also includes proper for Health Research and Development.
vector control. This can be done through State of the art: malaria research in the
environmental modification, biological control Philippines. Taguig: Department of Science
which includes using larvivorous fish in streams and Technology; 1999.
106 MedICal parasItology In the phIlIppInes

Bustos MD, Saul A, Salazar NP, Gorres NA. Myjak P, Nahorski W, Pieniazek NJ, Pietkiewitcz
Profile of Morong, Bataan, an area of low H. Usefulness of PCR for the diagnosis of
malaria endemicity in the Philippines. Acta imported malaria in Poland. Eur J Clinl
Trop. 1997;63:198–208. Microbiol Infect Dis. 2002;21(3):215–8.
Cabrera BD, Ramos OL, Cruz LT. Malaria Serghides L, Kain KC. Mechanism of protection
transmission by Anopheles littoralis, a salt induced by Vitamin A in falciparum
water breeder in Pangutaran, Sulu, Republic malaria. Lancet. 2002;359(9315):1404–6.
of the Philippines. Southeast Asian J Trop Warrell DA, Molyneux ME, Beales PF. Severe
Med Public Health. 1997;193–204. and complicated malaria. 2nd ed. Trans R
De Las Llagas LA. Malaria vectors and vector Soc Trop Med Hyg. 1990;81:521.
control. In: Philippine Council for Health World Health Organization. Antimalarial drug
Research and Development. State of the policies, report of an informal consultation.
art: malaria research in the Philippines. Geneva: World Health Organization; 1994.
Taguig: Department of Science and World Health Organization. Chemotherapy of
Technology; 1999. malaria and relapses and recrudescences:
Department of Health-National Epidemiology Report of a WHO scientific group.
Center. Field health service information Technical report series no. 529. Geneva:
system annual report 2008. Manila World Health Organization; 1993.
(Philippines): Department of Health; 2008. World Health Organization. Malaria diagnosis:
Department of Health. Administrative order new perspectives. Geneva: World Health
2009-0001: Revised policy and guidelines Organization; 2000.
on the diagnosis and treatment for malaria. World Health Organization. World malaria
Department of Health. Philippine malaria report 2009. Geneva: World Health
information system (PhilMIS) standard Organization; 2009.
operating guidelines. Manila (Philippines): World Health Organization. World malaria
Department of Health; 2009. report 2011. Geneva: World Health
Department of Health-National Center of Organization; 2011.
Disease Prevention and Control. Malaria World Health Organization. Management of
medium term development plan 2011– severe malaria: a practical handbook. 2nd
2016. Manila (Philippines): Department ed. Geneva: World Health Organization;
of Health; 2011. 2000.
Fidock DA. Drug discover y: Priming World Health Organization. Monitoring
the antimalarial pipeline. Nature. antimalarial drug resistance. Geneva:
2010;465:297–8. World Health Organization; 2001.
Hotez PJ. The “biblical diseases” and U.S. World Health Organization. Guidelines for
vaccine diplomacy. Brown World Aff J. the treatment of malaria. 2nd ed. Geneva,
2006; 12:247–58. Switzerland: World Health Organization;
Malaria Atlas Project. Clinical burden of 2010.
Plasmodium falciparum in 2010 globally. World Health Organization Western Pacific
2010 [cited 2012 Jun 5]. Available from Regional Office. Focus 2: Malaria, other
http://www.map.ox.ac.uk. vector borne and parasitic diseases. 2010
Medana LM, Day DP, Hien TT, Mai, NT, [cited 2012 Jun 5]. Available from: http://
Bethell D, Phu NH, et al. Axonal www.wpro.who
injury in cerebral malaria. Am J Pathol.
2002;60(2):655–66.
Chapter 2: protozoan Infections 107

Babesia spp.
Florencia G. Claveria

B abesia spp. is a hemosporidian parasite


that causes babesiosis, a hemolytic disease
commonly referred to as tick, splenic, redwater,
that infect red blood cells (RBCs) are generally
smaller and can easily be misidentified. Babesia
spp. are largely host-specific, and non-host
Texas, or Nantucket fever. Babesia was first specific species utilize only a narrow range
documented in cattle in 1888 by Dr. Victor of mammalian hosts like cattle and rodents.
Babes, a Romanian scientist, who described Some species infecting mammals exhibit cross-
symptoms of severe enzootic hemoglobinuria. infectivity and induce cross-immunity between
Babesia has a worldwide distribution and host species.
comprises approximately 73 to 100 species Hard ticks (family Ixodidae) are the
infecting wild and domestic animals, as well known biological hosts of Babesia, however,
as humans. Babesia spp. are generally specific transmission of Babesia by the soft tick,
to their vertebrate hosts, and are transmitted Ornithodoros erraticus has been reported. Tick
by Ixodidae or hard ticks. In humans, species that have been established as putative
transmission through blood transfusion, organ vectors are Boophilus spp., Rhipicephalus spp.,
transplantation, and transplacental route have Ixodes spp., Hyalomma spp., Haemaphysalis
been documented. spp., and Dermacentor spp. Of the 53 tick
As babesiosis affects a wide range of species incriminated as vectors, only 17 have
domestic animals, it undeniably brings about been recognized. The transmission to humans
huge economic losses to agriculture. The of the rodent B. microti and the cattle forms of
increasing association between man and animals Babesia is generally associated with Ixodes spp.
has resulted in increased infection, not only Unlike the genus Plasmodium and Theileria,
among the immunocompromised individuals Babesia does not undergo exo-erythrocytic
but also among the general populace. To merogony; daughter progeny are not housed in
increase public health awareness, a better parasitophorous vacuoles; and residual bodies
understanding of the parasite biology and are usually non-existent in infected RBCs.
its tick vector, the disease it causes, and its The Babesia life cycle undergoes three
epidemiology particularly in the Asia-Pacific, developmental phases. In the mammalian
is imperative. host, (a) merogony in the RBC; and in the tick
vector, (b) stages of gamogony in the gut and
Parasite Biology
epithelium; and, (c) sporogony accompanied
Babesia is a heteroxenous parasite requiring with multiple fission in various cells and organs
mammals as primary hosts, and ticks as forming sporokinetes, and the development of
intermediate hosts or vectors. On account of infective sporozoites. A few hours after blood
the disparity in the morphological features ingestion, the intra-erythrocytic merozoites in
of the intra-erythrocytic forms in different the gut of engorged ticks undergo morphologic,
host species, there exist about 100 species or physiologic/metabolic, and antigenic
forms. The tendency of Babesia spp. to take on changes, and differentiate into gametocytes
pleomorphic forms in different hosts obscure that eventually develop into gametes. Post-
their identification at the species level. For fertilization, the zygote begins to infect the
example, its close relative, the Theileria spp. gut epithelial cells where it undergoes multiple
108 MedICal parasItology In the phIlIppInes

fission, and eventually forms sporokinetes. Once the organs of the larva, nymphs, and adult
the sporokinetes are released, they continue to ticks. With the stage-to-stage (transstadial)
infect and multiply in various organs, including transmission, each of the developmental stages
the ovaries of the replete tick, until death ensues. is generally capable of parasite transmission to
The transovarian route represents one pattern mammals. The complicated phase of Babesia
of parasite transmission in the vector, which life cycle in the tick vector ends with the
terminates with the death of the vector. formation of the infective sporozoites in various
With the passage of sporokinetes to eggs organs or in the salivary glands, for subsequent
(transovarian), similar cycles of multiple fissions transmission to the mammalian hosts during
continue to take place in the embryo and in blood feeding (Figure 2.23).

Figure 2.23. Life cycle of Babesia spp.


(Accessed from www.dpd.cdc.gov/dpdx)

Pathogenesis and Clinical Manifestations bigemina (4-5 µm by 2-3 µm) and B. caballi (3
µm by 2 µm) are less virulent. Several factors can
Smaller forms like Babesia bovis (2.4 µm
influence the susceptibility of hosts to infection,
by 1.5 µm) and B. equi (2 µm by 1 µm) are
like the age and breed of farm animals, and the
more pathogenic, while larger forms such as B.
health and immune state of humans.
Chapter 2: protozoan Infections 109

Both innate and acquired immunity Diagnosis


contribute to the resolution of the primary
Babesia parasites are usually detectable in
infection and provide protection against
blood smears only during the acute stage of the
subsequent exposures. The importance of the
infection, and animals that survive the initial
spleen in the elimination of both the parasites
infection can become lifelong carriers. Previous
and infected RBCs is seen in the increased
infections can be demonstrated serologically.
susceptibility of splenectomized or inherently
Definitive diagnosis requires direct
asplenic mice to infection. Murine hosts
microscopic examination of Giemsa-stained
depleted of macrophages exhibit either high
peripheral blood smears for the presence
mortality or become unprotected when exposed.
of Babesia, showing its established unique
The transfer of primed macrophages, as opposed
morphological features. To rule out the
to transfer of primed T-cells, provides protection
misdiagnosis from closely related hemosporidians
against B. microti in naive mice. The resolution
like Plasmodium spp., and the causative agents
of babesiosis is principally mediated by gamma
of Lyme disease, and granulocytic erlichiosis
interferon produced by CD4+ T helper-1 cells,
infecting humans, the parasite dimensions
alongside macrophage activation. While B
and pleomorphism (ring form, pear-shaped,
cell-depleted mice are capable of controlling
and “Maltese cross” or tetrad form) need to be
primary infection, antibodies are still useful
ascertained, including the absence of pigments
in the clearance of extracellular parasites in
in infected RBCs. In cases of low-grade
circulation. Despite low parasitemia in the
infection or parasitemia, detection can be very
peripheral circulation among infected cattle,
difficult, thus, appropriate serological assays,
infected erythrocytes are sequestered in the
molecular gene analyses, and epidemiologic
capillary beds causing cerebral babesiosis, a
data, including data on ticks and reservoir
similar manifestation of falciparum malaria.
or carrier hosts (epizootiological), may be
In humans, infections with B. microti or
extremely useful.
B. microti-like species may be asymptomatic
Several serological tests are generally
or may result in mild to severe clinical signs
employed for the detection of babesiosis. The
and symptoms. Fatigue, malaise, anorexia, and
immunofluorescent assay (IFA) is widely used
weight loss begin to manifest approximately
in acute cases and in epidemiological studies.
one to six weeks post-exposure followed
Although sufficiently sensitive, IFA has the
by non-periodic or intermittent fever (38-
following drawbacks: non-differentiation
40°C), chills, and sweats accompanied
between active and past exposures, possibility
with headache, myalgia, arthralgia, nausea,
of cross-reactivity between antigens of
vomiting, and prostration. The patient may
closely related species, and subjectivity in the
also manifest emotional lability, depression,
quantification of the intensity of fluorescence.
and hyperesthesia. In severe cases, hemolytic
At lower dilutions and during the acute phase,
anemia and hemoglobinuria with jaundice
anti-B. microti antibodies cross-react with
become apparent, with pulmonary edema being
antigens of other Babesia spp. Also, antigens of
the most frequently observed complication
B. microti occasionally cross-react with sera of
of the disease. The severity of infection with
confirmed malarial cases with a >1:16 antibody
possible fatal outcome is generally associated
titer. To rule out possible cross-reactions, a 1:64
with the elderly, the splenectomized and
serum dilution is highly recommended. In cases
immunocompromised, and those manifesting
of low parasitemia, experimental inoculation of
evidence of Lyme disease.
specific pathogen-free hamsters with infected
110 MedICal parasItology In the phIlIppInes

blood or NOD/sch-scid mice with the patient’s The drug combination azithromycin and
blood can be useful in parasite detection and atovaquone is as effective as clindamycin-
identification. quinine, with less adverse effects. Both drug
The polymerase chain reaction (PCR) is combinations are ineffective in suppressing
highly specific and is generally considered to disease progression in immunosuppressed
be the gold standard for Babesia detection. It is, patients. Very recently, there have been reports
however, unrealistic for epidemiologic surveys of immunocompromised patients who,
because it is time consuming and expensive. during 28 days of uninterrupted treatment
The current practice is the use of PCR in the with azithromycin-atovaquone, manifested
isolation of the SSU rDNA from asymptomatic recurrence of marked parasitemia, suggesting
patients, followed by gene sequencing and its the development of drug-resistant B. microti.
comparison with known SSU rDNA gene Ar temisinin, pyrimethamine, and
sequences from pathogenic strains. pamaquine can strongly inhibit the growth of
The continued work on the isolation of B. equi and B. caballi in vitro. Pyrimethamine
specific and highly immunogenic antigens can indirectly interrupt the parasite life cycle
of Babesia species and isolates, and their through its inhibitory effect on dihydrofolate
intended utilization in the development reductase, essential in folate metabolism, while
of immunochromatographic test (ICT) is pamaquine can interfere in the recycling of
practicable for epidemiologic surveys in the endosomal proteins into the plasma membrane
field. ICT is simple, quick, reliable, sensitive, by direct interaction with the endosomes.
and inexpensive, and has gained acceptability in
Epidemiology
the diagnosis of both acute and latent infections.
Currently, there are ICT strips or dipsticks Babesiosis is essentially a zoonotic infection,
employed in the detection of infected livestock. regarded of major economic importance
to livestock, particularly in the cattle and
Treatment
horse industry. Its documentation in humans
The standard treatment of human babesiosis worldwide has increased its recognition as
utilizes a drug combination of clindamycin and a disease of public health concern. The first
quinine, or azithromycin and atovaquone. The human case attributed to the cattle form was
clindamycin and oral quinine combination reported in 1956, in Europe, and followed
was first used in 1982 in a newborn infant with reports of more cases in Europe and in
suffering from babesiosis, and since then, this North America, including the discovery of
combination has become the drug of choice. the transmission of a B. microti-like species to
Clindamycin is given 1.2 g intravenously twice humans in 1970. A review of the 136 human
a day or 600 mg orally three times a day, and cases examined in New York (1982-1991)
combined with oral quinine at a dose of 650 revealed almost all cases were among those
mg three times a day. Clindamycin-atovaquone living in Suffolk, Long Island. Of the 103
combination is efficacious in clearing the (76%) who were hospitalized, seven patients
parasites in normal individuals and prevents previously underwent splenectomy, 31 patients
recurrence of infection, but produces adverse had symptoms of babesiosis and Lyme disease,
effects like vertigo, tinnitus, and gastrointestinal and seven died. The Asian cases reported have
symptoms. Supportive and symptomatic been few and sporadic with the first records in
management is important. In severe cases where 1984, in Yunnan, China, and a recent report
there is progressive exacerbation of hemolytic in Japan attributed to B. microti (Table 2.10).
anemia, total blood exchange may be indicated.
Chapter 2: protozoan Infections 111

Table 2.10. Summary of human cases of babesiosis reported in some Asian countries

Location Signs and Symptoms Diagnosis


Yunnan, China Fever, jaundice, anemia, cutaneous edema; Initially malaria, then babesiosis
myalgia, malaise, nausea, prolonged and
repeated illness; periodic fever (39.5-41.0°C),
with renal transplantation prior to onset of fever
Taiwan Headache, malaise, fatigue, dull pain in upper Gallstone with hepatosplenomegaly
abdomen, and frequent mild fever, chill for a and babesiosis
few months, hemolytic anemia
Mongolia High fever, chill, nightly sweating, myalgia, low Babesiosis
grade parasitemia
Japan Fever, malaise, excretion of dark-colored urine, Babesiosis
progressive hemolytic anemia

The Centers for Disease Control and habitation with livestock and wild animals,
Prevention, USA has confirmed more than and where ticks were abundant. The parasites
40 human cases that contracted the disease detected were pyriform-shaped, suggestive of
from transfusion of packed RBC and tested Babesia. One case recorded in the rural area
positive for anti-B. microti antibodies. In Asia, in Southwestern Taiwan was serologically and
the two cases have been associated with renal morphologically diagnosed with a chronic and
transplantation and blood transfusion. Thus, subclinical infection of a geographic isolate
subclinical or asymptomatic cases cannot simply of Babesia named Taiwan isolate (TW1). The
be ignored, considering their potential role in detection of anti-Babesia antibodies in 83%
the spread of human babesiosis. Rattus coxinga endemic in the locality where the
Phylogenetic analyses of the gene sequences Taiwanese patient lived, suggested the rodents
of the SSU rDNA of Babesia spp. obtained as the highly likely source of infection. The SSU
from human cases helped clarify three patterns rDNA isolated from the Japanese patient and
or groupings, worldwide, namely: (a) human from the NOD/sch-scid mice inoculated with
babesiosis attributed to the B. divergens-related the patient’s blood revealed 99.2% sequence
parasites in Europe; (b) human babesiosis caused homology with the US B. microti SSU rDNA
by B. microti principally in the Northeastern (Genbank/EMBL/DDBJ: U09833).
USA; and (c) human babesiosis caused by newly In Japan the wild rodents, Apodemus
emerging species, the WA1-type in the Western speciosus and A. argenteus, are infected with B.
USA, tentatively grouped with B. microti or microti-like forms. In Taiwan, the bandicoot
alternatively with Theileria spp. Recently in rats, Bandicola indica, and the spiny rat, R.
Italy and Austria, parasites obtained from coxinga, carry morphologically similar B.
splenic cases revealed SSU rDNA sequences microti-like forms. The TW1 isolated from
more closely related to B. odocoileus, a species the first human case in Taiwan is serologically
that bears morphological, molecular, and related to the US B. microti SSU rDNA.
immunological similarities with B. divergens. In the Philippines, studies on animal
The B. divergens-related species now has been babesiosis have been limited and mainly
expanded to include B. odocoileus and possibly concentrated on hematological parameters and
B. bovis. In Asia, the etiologic agent of human clinical manifestations in cattle B. bigemina and
babesiosis has been identified as B. microti or B. argentina (syn. B. bovis), and B. canis. Using
B. microti-like isolate or strain. the ICT, 13 (28%) stray dogs in an impounding
Human cases recorded in China were facility in Dasmarinas, Cavite tested positive for
generally among farmers living in close anti-p50 truncated B. gibsoni antigen. The dogs
112 MedICal parasItology In the phIlIppInes

had infestation mainly with Rhipicephalus ticks, In Europe and the USA, the Ixodes ricinus
suggestive of their putative role in the spread of and Ixodes triangulicep, and the Ixodes scapularis
canine babesiosis in the country. and Ixodes pacificus are the principal vectors,
Slaughtered and race horses in the respectively. In Asia, the tick vectors are poorly
Philippines tested seropositive for B. caballi established. The predominance of Ixodes
and/or B. equi infection, using ICT containing a granulatus in Southeast Asian countries makes
recombinant B. caballi 48-kDa rhoptry protein it a favorable vector, though this warrants
(rBc48) and a recombinant truncated B. equi confirmation (Plate 2.17). In Japan, Ixodes
merozoite antigen 2 (rEMA-2t). Serological ovatus has been suggested as a highly likely
data correlated well with the detection of the tick vector of human babesiosis for its human
morphologies of the specific etiologic agent(s) biting activity.
in blood smears.

Plate 2.17. Ixodes sp. A. Non-engorged female. B. Engorged female. C. Mouthparts showing
the hypostome (H), pedipalp (P), and scutum (S). (Courtesy of Ms. Mary Jane Cruz-Flores
and Dr. Florencia Claveria)

Prevention and Control serological surveys reveal more of subclinical or


asymptomatic cases. Human cases of babesiosis
Babesiosis is essentially a zoonotic infection,
are generally associated with splenectomized
and its transmission to man through the
and immunocompromised patients, and
bite of the tick vector is incidental. Effective
noteworthy are the cases acquired through
prevention strategies include avoiding tick-
blood transfusion and organ transplantation.
infested areas, remaining covered with clothing,
There may be a need to consider the inclusion
and immediately removing any attached ticks.
of screening procedures for B. microti for blood
As the parasite is capable of stage-to-stage
and organ donors in high risk areas.
passage, each of the developmental stages is
capable of parasite transmission to mammals. References
The application of bug repellents in clothes
Allsopp MT, Allsopp BA. Molecular sequence
like N,N-diethyl-meta-toluamide (DEET)
evidence for the reclassification of some
and acaricides like permethrin may be useful.
Babesia species. Ann N Y Acad Sci.
The rodent population should be controlled as
2006;1081:509–17.
well, as they are major carriers or reservoirs of
Clark IA, Allison AC. Babesia microti and
the parasite.
Plasmodium berghei yoelli infections in
In low grade and chronic infections,
mice. Nature. 1974;252:328–9.
babesiosis is generally asymptomatic. Worldwide,
Chapter 2: protozoan Infections 113

Carlos ET, Carlos ER, Calalay FT, Cabiles He S, Saito-Ito A. Case reports of human
CC. Babesia canis: clinical observations in babesiosis from Asia. In: Yano A, Nam HW,
dogs in the Philippines. Phil J Vet Med. Anuar KA, Shen J, Saito A, Igarashi, I, editors.
1972;11:81–91. Toxoplasmosis and babesiosis in Asia. Vol
Clarke CR, Burrows GE, MacAllister 4, Asian Parasitology. Chiba (Japan): The
CG, Spillers DK, Ewing P, Lauer AK. Federation of Asian Parasitologists; 2005.
Pharmacokinetics of intravenously and p. 156–9.
orally administered pyrimethamine in Healy, G. and Ristic, M. Human babesiosis. In:
horses. Am J Vet Res. 1982;53:2292–5. Ristic M, editor. Babesiosis of Domestic
Cruz-Flores MJ, Claveria FG, Verdida R, Xuan Animals and Man. Boca Raton, Florida:
X, Igarashi I. First detection of Babesia CRC Press; 1988. p. 209–28.
gibsoni infection in Philippine stray dogs Herwaldt BL, Caccio S, Gherlinzoni F, Aspock
by immunochromatographic test (ICT). H, Slemenda SB, Piccaluga P, Martinelli
Veterinarski Arhiv. 2008;78:149–57. G, et al. Molecular characterization of a
Cruz-Flores MJ, Bata M, Co B, Claveria non-Babesia divergens organism causing
F G , Ve r d i d a R , Xu a n X , e t a l . zoonotic babesiosis in Europe. Emer Infect
Immunochromatographic Assay of Babesia Dis. 2003;9:942–8.
caballi and Babesia equi Laveran 1901 Igarashi, I., Suzuki, R., Waki, S., Tagawa, Y.,
(Theileria equi Mehlhorn and Schein, Seng, S. Tum, S., et al. Role of CD4+ T
1998) (Phylum Apicomplexa) infection in cells and gamma interferon in protective
Philippine horses correlated with parasite immunity against Babesia microti infection
detection in blood smears. Veterinarski in mice. Infect Immun. 1999;76:4143–8.
Arhiv. 2010;80:715–22. Krause PJ, Lepore T, Sikand VK, Gadbaw
Dantrakool A, Somboon P, Hashimoto T, J Jr, Burke G, Telford SR III., et al.
Saito-Ito A. Identification of a new type Atovaquone and azithromycin for the
of Babesia species in wild rats (Bandicota treatment of babesiosis. N Engl J Med.
indica) in Chiang Mai Province, Thailand. 2000;343:1454–8.
J Clin Microbiol. 2004;42:850–4. Krause PJ, Spielman A, Telford SR III, Sikand
Dumag PU, Reyes PV. Occurrence of VK, McKay K, Christianson D, et al.
endoglobular parasites morphology Persistent parasitemia after acute babesiosis.
resembling Babesia argentina in Santa N Engl J Med. 1998;339:160–5.
Gertrudes bulls and Shorthorn cattle at the Kuttler KL. Worldwide impact of babesiosis.
Bongabon stock farm quarantine station, In: Ristic M, editor. Babesiosis of Domestic
Bongabon, Nueva Ecija. Phil J Anim Ind. Animals and Man. Boca Raton, Florida:
1960;21:221–4. CRC Press; 1988. pp. 1–22.
Friedhoff KT. Transmission of Babesia. In: Ristic Li JF, Meng DB, Wang QF, Dong XG, Wang
M, editor. Babesiosis of Domestic Animals JY. The discovery of human babesiosis.
and Man. Boca Raton, Florida: CRC Press; Chinese J Vet Med. 1984;10:19–20.
1988. p. 23–54. Lux JC, Weiss D, Linden JV, Kessler D, Herwaldt
Goodger BV, Rood-Bramanis K, Wright BL, Wong SJ, et al. Transfusion-associated
IG. Procoagulant activity of Babesia babesiosis infection after heart transplant.
bovis-infected erythrocytes. J Parasitol. Emer Infect Dis. 2003;9(1):116–9.
1987;73:1052-3. Manwell DR, Kuntz ER. A new Babesia
Gunders AE. Piroplasmal sporozoites in the from the Indian bandicoot. J Parasitol
argasid Ornithodoros erraticus (Lucas). 1964;50:390–3.
Experientia. 1977;33:892–3.
114 MedICal parasItology In the phIlIppInes

Meldrum SC, Birkhead GS, White DJ, Benach Toxoplasmosis and babesiosis in Asia. Vol
JL, Morse DL. Human babesiosis in New 4, Asian parasitology. Chiba (Japan): The
York: an epidemiological description of 136 Federation of Asian Parasitologists; 2005.
cases. Clin Infect Dis. 1992;15:1019–23. p. 160–9.
Meeusen E, Lyod S, Soulsby EJ. Babesia microti Saito-Ito A. Introduction: Human babesiosis in
in mice: Adoptive transfer of immunity the world. In: Yano A, Nam HW, Anuar
with serum and cells. Aust J Exp Biol Med K, Shen J, Saito A, Igarashi I, editors.
Sci. 1984;62:551–66. Toxoplasmosis and babesiosis in Asia. Vol
Mills CD, Burgess DC, Taylor HJ, Kain KC. 4, Asian parasitology. Chiba (Japan): The
Evaluation of a rapid and inexpensive Federation of Asian Parasitologists; 2005.
dipstick assay for the diagnosis of p. 145–54.
Plasmodium falciparum malaria. Bull World Shih CM, Wang CC. Ability of azithromycin
Health Organ. 1999;77:553–9. in combination with quinine for the
Mohebale M, Taran M, Zarei Z. Rapid elimination of babesial infections in humans.
detection of Leishmania infantum infection Am J Trop Med Hyg. 1998;59:509–12.
in dogs: comparative study using an Shi ZB, Li ZZ, Gao QR, Ding YM. A case
immunochromatographic dipstick rk39 of human babesiosis. Chinese J Parasitol
test and direct agglutination. Vet Parasitol. Parasitic Dis. 1996;14:240.
2004;121:239–45. Shih CM, Liu LP, Chung WC, Ong SJ,
Molina JM, Montenegro MM. Anaplasmosis Wang CC. Human babesiosis in Taiwan:
in two Philippine cows. Phil J Vet Med. asymptomatic infection with a Babesia
1977;16:181–7. microti-like organism in a Taiwanese
Nagai A, Yokoyama N, Matsuos T, Bork S, woman. J Clin Microbiol. 1997;35:450–4.
Xuan X, Zhu Y, et al. Growth inhibitory Su GG, Zhao NF, Ye YX. A case report
effects of artesunate, pyrimethamine, and of babesiosis. Chinese J Zoonoses.
pamaquine against Babesia equi and Babesia 2002;18:112.
caballi in in vitro cultures. Antimicrob St. John JH, Simmons JS, Reynolds FH.
Agents Chemother. 2003;47:800–3. A survey of mongrel dogs in Manila,
Okamura M, Igarashi I. Host Immune Philippine Islands, for helminthic and
responses against Babesia infection. In: blood protozoan parasites. Phil J Sci.
Yano A, Nam HW, Anuar K, Shen J, Saito 1931;44:309–11.
A, Igarashi I, editors. Toxoplasmosis and Tadaka N, Yano Y. Vector ticks for human
babesiosis in Asia. Vol 4, Asian parasitology. babesiosis in Asia. In: Yano A, Nam HW,
Chiba (Japan): The Federation of Asian Anuar K, Shen J, Saito A, Igarashi I, editors.
Parasitologists; 2005. p. 1224–32. Toxoplasmosis and babesiosis in Asia. Vol
Saito-Ito A, Tsuji M, Wei Q, He S, Matsui T, 4, Asian parasitology. Chiba (Japan): The
Kohsaki M, et al. Transfusion-acquired Federation of Asian Parasitologists; 2005.
autochthonous human Babesiosis in Japan: p. 170–3.
isolation of Babesia microti-like parasites Telford SR, Gorenflot A, Brasseur P, Spielman
in hu-RBC-SCID mice. J Clin Microbiol. A. Babesia infection in humans and wildlife.
2000;38:4511–6. In: Kreir JP, editor. Parasitic protozoan. Vol
Saito-Ito A, Dantrakool A. Babesia microti-like 5. San Diego, California: Academic Press;
parasites in Asia: as potential parasites for 1993. p. 1–47.
human babesiosis. In: Yano A, Nam HW, Van Weert AW, Geuze HJ, Groothuis B,
Anuar K, Shen J, Saito A, Igarashi I, editors. Stoorvogel W. Primaquine interferes with
Chapter 2: protozoan Infections 115

membrane recycling from endosomes to of resistance to azithromycin-atovaquone


the plasma membrane through a direct in immunocompromised patients with
interaction with endosomes, which does Babesia microti infection. Clin Infect Dis.
not involve neutralization of endosomal 2010;50:381–6.
pH or osmotic swelling of endosomes. Eur Yamane I, Thomford JW, Gardner IA, Dubey
J Cell Biol. 2000;79:394–9. JP, Levy JP, Conrad PA. Evaluation of
Van Peenen PF, Chang SJ, Banknieder AR, the indirect fluorescent antibody test for
Santana FJ. Piroplasm from Taiwanese diagnosis of Babesia gibsoni infections in
rodents. J Protozool. 1977;24:310–2. dogs. Am J Vet Res. 1993;54:1579–84.
Wittner M, Rowin KS, Tanowitz HB, Hobbs Zamoto A, Tsuji M, Wei Q, Cho SH, Shin EH,
JF, Saltzman S, Wenz B, et al. Successful Kim TS, et al. Epizootiologic survey for
chemotherapy of transfusion babesiosis. Babesia microti among small wild mammals
Ann Int Med. 1982;96:601–4. in northeastern Eurasia and a geographic
Wormser GP, Prasad A, Neuhaus E, Joshi S, diversity in the beta-tubulin gene sequences.
Nowakowski J, Nelson J, et al. Emergence J Vet Med Sci. 2004;66:785–92.
116 MedICal parasItology In the phIlIppInes

Blood and Tissue Flagellates


Vicente Y. Belizario, Jr., Julius R. Migriño, Jr.

L ocally acquired infections due to the blood


and tissue flagellates have not yet been
documented in the Philippines. However,
get infected include the skin, gonads, intestinal
mucosa, and placenta.
It has been well established that the
because of fast and easy travel, as well as arthropod vector responsible for propagating
increased human migration, imported cases this parasite are the reduviid bugs, belonging
from endemic countries may become future to the genera Triatoma, Panstrongylus, and
sources of local infection. This scenario is Rhodnius. These arthropods thrive under
possible because the vectors of Trypanosoma squalid housing conditions such as thatched
cruzi, Triatoma and Rhodnius bugs, are found roofs and mud walls, commonly seen in poor
in the country. In the same manner, the rural communities. Zoonotic mammalian
Philippines has a number of Phlebotomus spp., reservoir hosts have been identified, including
which can serve as vectors for Leishmania spp. domestic animals, armadillos, raccoons,
rodents, marsupials, and even some primates.
Trypanosoma cruzi T. cruzi exhibits all four stages of
development: amastigote, promastigote,
Trypanosoma cruzi is the etiologic agent of epimastigote, and trypomastigote. In humans,
Chagas disease or American trypanosomiasis. trypomastigotes are found in the bloodstream,
This is the only parasite that was discovered and amastigotes in tissue cells (Figure 2.24).
and studied before it was known to cause a Inside its insect vector, the amastigote,
disease. More than 100 years ago, Carlos Chagas epimastigote, and promastigote forms occur
found that the trypanosomes he dissected from in the midgut, while the infective metacyclic
the intestine of a triatomid bug were the same trypomastigote appear in the hindgut.
parasites found in the blood of a child suffering Amastigotes are round or ovoid in shape
from fever and enlargement of the lymph nodes. and measure from 1.5 to 4 µm in diameter. They
Since then, the understanding behind the are usually found in small groups of cyst-like
disease that this protozoan causes has shown collections in tissues.
profound changes in terms of pathophysiology, The long slender trypomastigotes are 16
diagnosis, and treatment. to 20 µm in length while the short, stumpy
Parasite Biology forms measure around 15 µm (Plate 2.18).
The posterior end is usually pointed. The
T. cruzi belongs to the trypanosome undulating membrane is narrow with two to
group Stercoraria. Trypanosomes under this three undulations, and a single thread-like
group multiply within the mammalian host flagellum originating near the kinetoplast
in a discontinuous manner. Unlike other provides the parasite with mobility. In stained
trypanosomes, it is an intracellular parasite, with specimens, trypomastigotes are characteristically
myocytes (particularly myocardial tissues) and C-shaped. They have also been described as
cells of the reticuloendothelial system being the U- or S-shaped with a prominent kinetoplast,
most heavily infected cells. Other tissues that characteristic of the species.
Chapter 2: protozoan Infections 117

Figure 2.24. Life cycle of Trypanosoma cruzi


(Accessed from www.dpd.cdc.gov/dpdx)

The trypomastigotes of T. cruzi do not


multiply in the bloodstream. Soon after their
entry into the human host, the metacyclic
trypanosomes are engulfed by macrophages
of the reticuloendothelial system and multiply
through binary fission as amastigotes.
Amastigotes develop into trypomastigotes,
and the cells lyse in 4 to 5 days. The released
trypomastigotes enter the bloodstream, ready to
replicate again once they enter another cell or
are ingested by an insect vector. Once ingested
by the intermediate host, the trypomastigotes
pass through the posterior portion of the
insect’s midgut and become epimastigotes,
where they multiply via longitudinal fission.
Infective metacyclic trypomastigotes appear
Plate 2.18. Trypanosoma cruzi trypomastigote in
thin blood smears stained with Giemsa
in the insect’s rectum 8 to 10 days after
(Accessed from www.dpd.cdc.gov/dpdx) infection, and are passed in the bug’s feces.
The metacyclic trypomastigotes gain entry
118 MedICal parasItology In the phIlIppInes

into the body through broken skin, or through result in cardiomegaly, congestive heart failure,
mucous membranes that are rubbed with fingers thromboembolism, and even arrhythmias. Less
contaminated with the bug’s feces. severe signs and symptoms associated with
the chronic phase of the disease include chest
Pathogenesis and Clinical Manifestations
pain, palpitations, dizziness, syncopal episodes,
Chagas disease can be divided into an abnormal electrocardiogram findings, achalasia
acute and a chronic phase. The acute phase is associated with megaesophagus, and chronic
characterized by a focal or diffuse inflammation constipation associated with megacolon. About
mainly affecting the myocardium. Non- one-third of patients in the latent stage develop
specific signs and symptoms, such as fever, some manifestation of chronic Chagas disease
malaise, nausea, vomiting, and generalized after several years or decades. The majority of
lymphadenopathy often accompany the acute symptomatic, chronic patients manifest with
phase. Cutaneous manifestations of the disease the cardiac form, while the rest develop the
can sometimes appear during this phase, gastrointestinal form.
usually associated with a localized inflammatory
Diagnosis
reaction at or near the site of inoculation.
Chagomas are furuncle-like lesions associated A complete patient history is the primary
with induration, central edema, and regional tool for diagnosing Chagas disease. Possible
lymphadenopathy. These lesions represent exposure to T. cruzi should be established, and
the site of entry of the parasite. If the parasite risk factors such as place of residence or work,
penetrates through the conjunctiva, eyelid recent blood transfusion in an endemic area,
swelling called Romaña’s sign may form. This and contact or exposure to T. cruzi intermediate
lesion is characterized by unilateral painless host should be evaluated.
bipalpebral edema and conjunctivitis, and may The definitive diagnosis of Chagas
involve the lacrimal gland and surrounding disease during its acute phase relies on direct
lymph nodes. After 1 or 2 months, symptoms visualization of the parasites in thick and thin
resolve, and the patient goes into a latent or blood smears using Giemsa stain. Cerebrospinal
indeterminate, but usually asymptomatic phase. fluid (CSF), tissue samples, or lymph can also be
During this phase, patients infected with T. used for parasite visualization. However, only in
cruzi are still capable of transmitting it to others the first two months of acute disease can T. cruzi
through insect vectors, blood transfusion, or trypomastigotes be seen by direct examination.
organ transplantation. Other diagnostic techniques include
The pathophysiology of the chronic concentration methods (microhematocrit),
phase of the disease was initially thought to blood culture, and polymerase chain reaction
be autoimmune in nature; however, this is (PCR). Xenodiagnosis, wherein laboratory-
controversial. Newer evidence shows that reared triatomine bugs are allowed to feed on
chronic Chagas disease is multifactorial, suspected patients and are later examined for the
and dependent on the interaction between presence of T. cruzi metacyclic trypomastigotes,
parasite and host. Nonetheless, the chronic may also be utilized as a diagnostic modality.
phase is manifested by fibrotic reactions that During the chronic phase, a variety of
cause injury to the myocardium, cardiac serologic tests may be used, such as enzyme-
conduction network, and enteric nervous linked immunosorbent assay (ELISA), indirect
system (decrease in nerve ganglia leading to hemaglutination, indirect immunofluorescence,
megasyndromes). The heart is the primary and PCR. The WHO recommends using at
organ affected during this phase, which may least two techniques with concurrent positive
Chapter 2: protozoan Infections 119

results before a diagnosis of Chagas disease The prevalence and distribution of


is made. For the cardiac form of the disease, American trypanosomiasis has been continually
ECG and echocardiography may show atrial shifting. Endemic regions include most of
fibrillation/flutter, low QRS voltage, dilated Central America and the southern cone of South
cardiomyopathy, and tricuspid and mitral America. Several factors tend to determine the
regurgitation. The gastrointestinal form is changes in prevalence of the disease in endemic
usually diagnosed by barium esophagogram countries. In certain regions in Mexico, delayed
(esophageal dilatation) and barium enema control policies and mobilization probably
(megacolon of the sigmoid and rectum). contributed to an increase in prevalence. In
the past 25 years, there have been several
Treatment
international efforts to control and prevent the
Two drugs are recommended for the disease in these endemic areas, most notably
treatment of acute phase Chagas disease: vector control strategies, and improved blood
nifurtimox and benznidazole. These drugs are transfusion safety regulations. Brazil, Chile,
usually associated with severe side effects: (a) Uruguay, and several areas of Argentina and
nifurtimox may cause weight loss, anorexia, Paraguay have eliminated the vector Triatoma
behavioral changes, and an antabuse effect; (b) infestans. However, the disease persists due to
benznidazole may cause rashes, bone marrow emergence of secondary domestic vectors, and
suppression, and peripheral neuropathy. vector resistance to insecticides.
Other classes of drugs, such as allopurinol Initially thought to be confined within the
and itraconazole, have been shown to halt the Latin American region, countries such as USA,
progression into cardiomyopathy, although no Canada, Spain, France, Switzerland, Japan, and
form of treatment has been shown to reverse Australia have seen a number of cases, primarily
damage caused by the disease. Newer drugs due to human migration patterns as well as from
such as triazole derivatives (posaconazole, blood transfusion, organ donation and vertical
ravuconazole) and cruzipain inhibitors transmission. However, these countries are still
(a parasite protease) are currently under regarded as non-endemic, and majority of the
development. cases are attributed to imported infections from
Symptom-specific management is used endemic areas.
to treat patients with chronic manifestations. Chagas disease is included in the WHO list
Cardiac manifestations are controlled by of Neglected Tropical Diseases (NTDs), and is
pacemakers and antiarrhythmic drugs, such the leading cause of parasite-related deaths in
as amiodarone, while megasyndromes are Latin America. In 2003, it ranked 3rd as the
managed with special diets, laxatives, and leading cause of parasitic infection in the world,
surgical procedures. behind malaria and schistosomiasis.
Epidemiology Prevention and Control

Chagas disease is estimated to have infected There have been major breakthroughs
more than 10 million people worldwide. Most in the control and prevention of American
cases are reported in the Latin Americas, where trypanosomiasis, particularly by Brazil, Chile,
more than 25 million people are at risk for the and Uruguay. Vector control and blood
disease. Serologic techniques have identified that transfusion regulations have delivered positive
up to 13% of populations in certain endemic outcomes, in terms of disease prevention in
regions are positive for T. cruzi antibodies. An these countries. Spraying of insecticides, use
estimated 10,000 to 12,000 people die of the of insecticide-treated bed nets, and house
disease annually. improvements to prevent vector infestation
120 MedICal parasItology In the phIlIppInes

have been proven cost-effective. International highly fatal disease caused by two subspecies of
organizations such as the WHO and the Trypanosoma brucei: T. brucei gambiense and T.
manufacturers of the antiparasitic drugs are brucei rhodesiense. A third subspecies, T. brucei
working in tandem to ensure the availability of brucei, primarily affects wild and domestic
drugs for the treatment of the disease. animals; collectively, the three subspecies
In Mexico and non-endemic countries represent the Trypanosoma brucei complex. The
near endemic countries, the coverage, quality earliest reports of sleeping sickness in Africa
and safety of blood transfusion screening date back to 1734, while the formal correlations
is being evaluated as avenues for disease between the symptoms, the parasite in blood
prevention. Vaccine development has not and CSF, and the relationship between the
yet been successful, but the advent of newer parasite and its insect vector were established
technologies and characterization of the T. cruzi during the early 1900s.
genome may aid in future vaccine research.
Parasite Biology
Trypanosoma brucei gambiense Members of the T. brucei complex belong to
Trypanosoma brucei rhodesiense the trypanosome family Salivaria. The parasite
is usually transmitted via the bite of the blood-
Human African trypanosomiasis (HAT), sucking tsetse fly (Glossina spp.) feeding from an
also known as African sleeping sickness, is a infected mammalian host (Figure 2.25). Since

Figure 2.25. Life cycle of Trypanosoma brucei


(Accessed from www.dpd.cdc.gov/dpdx)
Chapter 2: protozoan Infections 121

the disease relies heavily on the tsetse fly for its through mechanical methods (accidental needle
transmission, HAT cases are localized in regions pricks, other blood sucking insects), as well as
of sub-Saharan Africa, primarily in remote rural vertically, via mother-to-child infection through
areas where tsetse fly habitats are located. the placenta.
T. brucei gambiense is localized mostly in
Pathogenesis and Clinical Manifestations
the western and central regions of sub-Saharan
Africa. It primarily affects humans, but utilizes Human African trypanosomiasis has two
dogs, pigs, and sheep as reservoir hosts. It is types, acute and chronic, depending on the
responsible for the chronic type of sleeping subspecies causing the disease. Trypanosoma
sickness, and accounts for 95% of all HAT cases. brucei gambiense sleeping sickness manifests
T. brucei rhodesiense is found in east Africa months or years after initial infection, while
and is primarily a zoonosis of cattle and wild symptoms of T. brucei rhodesiense sleeping
animals, with humans being accidental hosts. sickness may appear just weeks after infection.
It causes the more acute and rapidly fatal form The initial lesion of African trypanosomiasis
of sleeping sickness, and accounts for the begins as a local, painful, pruritic, erythematous
remaining 5% of HAT cases. chancre located at the bite site, progressing
Only the epimastigote and trypomastigote into a central eschar, and resolving after 2 to
forms are exhibited by the T. brucei complex. 3 weeks. This trypanosomal chancre is more
The trypomastigotes are polymorphic: there common in Gambian sleeping sickness. Several
are typical slender forms, and short, stumpy days after the development of the chancre,
forms. They are flattened and fusiform in usually within 3 to 10 days, the next stages of
shape, 14 to 33 µm in length and 1.5 to 3.5 the disease manifest.
µm in width. The body tapers anteriorly and is Both types of HAT undergo two stages:
blunt posteriorly. The centrally located nucleus early and late. During the early phase of HAT,
contains a large central karyosome. There is an called the hemolymphatic stage, the parasites
undulating membrane, and a single flagellum proliferate in the bloodstream and lymphatics.
that runs along the edge of the undulating The patient may manifest with irregular bouts
membrane and becomes free anteriorly. of fever, headache, joint and muscle pain, and
Once ingested by the intermediate malaise. Anemia, myocardial inflammation,
host, Trypanosoma brucei trypomastigotes disseminated intravascular coagulation, and
undergo several developmental changes from renal insufficiency may occur. Frequently,
trypomastigote into procyclic forms in the in Gambian trypanosomiasis, the posterior
insect’s midgut. After multiplying for 15 to 20 cervical lymph nodes are enlarged, non-tender,
days, the epimastigotes migrate to the foregut and rubbery in consistency (Winterbottom’s
into the insect’s salivary glands, where they sign). Other lymph nodes, such as axillary
mature into metacylic trypomastigotes. When and supraclavicular lymph nodes, may also be
the infected fly bites another mammalian host, involved in both types of sleeping sickness. The
these infective trypomastigotes are injected into signs and symptoms manifested within this
the new host where they multiply and mature phase are due to tissue damage, either from
in blood and connective tissue. In humans, parasitic toxins or immune complex reactions
T. brucei lives in the blood, in the reticular that target organs and RBCs. The early systemic
tissue of lymph and spleen, and the CSF. The phase lasts from 1 to 6 months.
long, slender trypomastigotes multiply by The late phase of the disease, known as
longitudinal binary fission. the meningoencephalitic stage, marks the
Though mostly transmitted through its involvement of the central nervous system.
insect vector, the disease can also be transmitted The brain and meninges become involved
122 MedICal parasItology In the phIlIppInes

as the parasites find their way into the CNS due the relative higher levels of parasitemia.
through the bloodstream. This usually occurs Serial examinations may be necessary due
3 to 10 months after initial infection in to varying levels of parasitemia. Other
Gambian infections, but can manifest after diagnostic techniques include enzyme-linked
just a few weeks in Rhodesian trypanosomiasis. immunosorbent assay, immunofluorescence,
Neurologic symptoms become evident, such indirect hemagglutination test, mini-anion
as apathy, behavioral changes, headache, and exchange centrifugation technique, and PCR.
sleep pattern changes. These may be followed CSF examination is mandatory in patients
by more severe symptoms, such as convulsions, with suspected HAT to detect CNS involvement.
tremors, speech defects, disturbances in speech Abnormal CSF findings include increase in cell
and reflexes, and even paralysis. Kerandel’s sign count, opening pressure, protein concentration,
may manifest as a deep, delayed hyperesthesia and IgM levels. The latter is considered
(delayed bilateral pain out of proportion to pathognomonic for the meningoencaphalitic
the extent of tissue injury). In the later stages, stage of the disease.
somnolence manifests, followed by a deep coma. Card agglutination test for trypanosomiasis
Death eventually follows either from the disease (CATT) detecting circulating antigens in
itself, or from intercurrent infection due to persons infected with T. brucei complex is
immunosuppression. available commercially and can be used in the
Areas of the CNS usually involved in the field setting to screen at-risk populations. This
meningoencaphalitic phase include the frontal technique provides a rapid and highly specific
lobes, pons, medulla, and perivascular areas. method of screening for HAT cases; however,
Parasites may also be seen in the CSF. Autopsy the method has low sensitivity for certain strains
of HAT patients reveals edema and numerous, of T. brucei gambiense in certain areas of West
small, and confluent hemorrhages. Africa.
Trypanosomes are able to evade the immune
Treatment
response of the host through a process called
antigenic variation. This refers to the ability Treatment of African sleeping sickness
of the trypomastigote to continuously change depends on the stage of the disease. For the first
its surface coat, composed of variant surface stage, intravenous suramin sodium for both T.
glycoproteins, so that the host’s antibodies brucei gambiense and T. brucei rhodesiense, and
cannot recognize the parasite in subsequent intramuscular pentamidine for the Gambian
recurrent waves of parasitemia. form can be used. These drugs have side effects,
which include fever, rash, renal insufficiency,
Diagnosis
muscle pain, and paresthesia for suramin; and
Diagnosis of human African trypanosomiasis tachycardia, hypotension, and hypoglycemia
depends upon the demonstration of highly for pentamidine. These drugs do not cross the
motile trypomastigotes in expressed fluid from blood-brain barrier, and so, they cannot be used
a chancre, lymph node aspirate, and CSF. for the CNS stage of the disease.
Thick and thin blood films can be stained Once CNS involvement occurs, intravenous
with Giemsa. Buffy coat concentration method melarsoprol is the drug of choice for both types
is recommended to detect parasites when of sleeping sickness. This arsenic-containing
they occur in low numbers. Examination for drug can cause fatal arsenic encephalopathy
trypomastigotes is usually done during the (usually prevented by co-administration of
hemolymphatic stage of the disease, and is more corticosteroids), and resistance to the drug
useful for the diagnosis of T. brucei rhodesiense has also been observed. A febrile episode
Chapter 2: protozoan Infections 123

called a Jarisch-Herxheimer reaction due do their laundry. Rhodesian trypanosomiasis


to trypanosome lysis may occur following is an occupational hazard for persons working
melarsoprol treatment. in game reserves, and may also be a threat to
A second-line drug, nitrofurazone, is used visitors of game parks. Cattle and game animals
in cases of melarsoprol treatment failure. A like antelopes can serve as reservoir hosts for
newer drug, eflornithine, is less toxic than the parasite.
melarsoprol, and can also be used during
Prevention and Control
the hemolymphatic stage; however, it is only
effective against T. brucei gambiense. Recent Vector control is the primary method used
evidence has shown that a combination in the control and prevention of African sleeping
treatment of oral nifurtimox and intravenous sickness. Tsetse fly trapping is the main strategy
eflornithine is of similar efficacy compared to employed to decrease the vector population.
longer intravenous monotherapy with either Use of insecticides and protective clothing are
agent. Combination therapy is advantageous recommended to prevent contact with the insect
due to the relative ease in administration, and vector. Regulation and treatment of reservoir
a decreased risk of developing drug resistance. hosts such as cattle and game animals are also
Although nifurtimox is currently registered as being looked upon as an effective means of
a drug against American trypanosomiasis, its preventing disease transmission.
use in the nifurtimox-eflornithine combination Several programs developed to eliminate the
treatment (NECT) has been included in the insect vector have been in place in Africa. The
WHO List of Essential Medicine. Kwando-Zambesi Regional Tsetse Eradication
project started in Botswana, and in 2000, the Pan
Epidemiology
African Tsetse and Trypanosomiasis Eradication
Sleeping sickness affects around 300,000 Campaign (PATTEC) was established. Aerial
to 500,000 people in 36 countries within sub- and localized spraying of insecticides in Angola,
Saharan Africa. It is estimated that more than Botswana, Namibia, and Zambia has eradicated
50 million people are at risk of infection. In the the tsetse fly in these African countries.
last 10 years, most reported cases came from The WHO has established partnerships
the Democratic Republic of Congo (DRC), with private companies such as Aventis Pharma
followed by the Central African Republic. (Sanofi-Aventis) and Bayer HealthCare to
Other countries such as Angola, Cameroon, provide surveillance and management support
Chad, Congo, Côte d’Ivoire, Equatorial Guinea, to endemic countries.
Gabon, Guinea, Kenya, Malawi, Nigeria,
Sudan, Uganda, United Republic of Tanzania, Leishmania spp.
Zambia, and Zimbabwe have also reported
Early descriptions of leishmaniasis have been
cases.
found as early as the first century A.D., where
During the turn of the century, between
American Indians documented the disease in
10,000 and 40,000 annual cases of HAT were
pottery figures. Cunningham studied the “Delhi
being reported. However, newer data from the
boil” in India back in 1885, and Leishman had
WHO has estimated more recently that new
properly identified the intracellular parasites in
cases have dropped below the 10,000 mark, a
1903. Leishmania braziliensis was later identified
first in 50 years.
in 1911 by Gaspar Viana, as was the insect
Tsetse flies live near the banks of rivers and
vector which transmitted the parasite in 1922
streams, therefore transmission can readily occur
by Henrique Aragao.
when people frequent these areas to swim and
124 MedICal parasItology In the phIlIppInes

Parasite Biology Mexico, Central America, and some parts of


South America, as well as the Amazon rain
Leishmaniasis is a disease caused by infection
forest, and is usually caused by L. mexicana, L.
of the diploid protozoa belonging to the genus
amazonensis, L. guyanensis, L. braziliensis, and L.
Leishmania. This genus is actually divided into
chagasi. Arthropods, particularly sandflies of the
two subgenera, differentiated from one another
genera Phlebotomus (Old World) and Lutzomyia
by the location of their development inside
(New World), act as the insect vector for these
the insect vector, as well as the areas in which
parasites. Dogs are the primary reservoir in
they are endemic. Currently there are about
urban areas, and rodents also act as reservoirs
15 species of Leishmania which cause clinical
in both urban and rural areas.
manifestations in humans. This diverse pool
Leishmania spp. produce amastigotes
of different species is historically divided and
intracellularly in the mammalian host, and
classified based on their biological, clinical,
promastigotes in the hindgut (Viannia subgenus),
geographic, and epidemiological characteristics.
midgut (Viannia and Leishmania subgenera),
Epidemiologically, the Leishmania spp.
and proboscis (Viannia and Leishmania
are divided into Old World and New World
subgenera) of the insect vectors. Amastigotes are
leishmaniasis. In the Old World, the common
ovoid or rounded bodies measuring 2 to 3 µm
species involved are L. tropica (Asia and
in length and live intracellularly in monocytes,
Eastern Europe), L. aethiopica (Africa) and
polymorphonuclear leukocytes, or endothelial
L. major. New World leishmaniasis affects
cells. The nucleus is large, while an axoneme

Figure 2.26. Life cycle of Leishmania spp.


(Accessed from www.dpd.cdc.gov/dpdx)
Chapter 2: protozoan Infections 125

arises from the kinetoplast and extends to the incubation period ranges from two weeks to
anterior tip. several months. An erythematous papule or
Promastigotes have a single free flagellum nodule, called an “oriental button,” is produced
arising from the kinetoplast at the anterior end. at the inoculation site. The lesion has raised
They measure 15 to 20 µm in length and 1.5 to edges and a central crater. During the course
3.5 µm in width. The infective promastigotes in of several weeks, the papule forms a violaceous
the proboscis of the sandfly are injected into the ulcer as it enlarges in size. The lesion may heal
host’s skin during feeding (Figure 2.26). They spontaneously after a few months, leading to
then invade the cells of the reticuloendothelial a disfiguring scar; in the case of New World
system, transform into amastogotes, and leishmaniasis, CL may progress to other forms
multiply via binary fission. When the parasitized of leishmaniasis.
cell ruptures, the amastigotes that are released
B. Diffuse Cutaneous Leishmaniasis
either invade new cells, or are taken up by
sandflies during feeding, where they transform The manifestation of DCL, also called
into promastigotes in the gut, multiply by anergic or lepromatous leishmaniasis, is
binary fission, and migrate to the foregut. characterized by a localized, non-ulcerating
Leishmania spp. may also be transmitted papule, eventually developing numerous
congenitally, through blood transfusion, by diffuse satellite lesions that affect the face and
contamination of bite wounds, and by direct extremities. This type of leishmaniasis may be
contact with contaminated specimens. initially diagnosed as lepromatous leprosy.
Pathogenesis and Clinical Manifestations C. Mucocutaneous Leishmaniasis

Clinically, leishmaniasis can be divided Mucocutaneous leishmaniasis develops


into four categories: cutaneous leishmaniasis in about 2 to 5% of persons infected with L.
(CL), diffuse cutaneous leishmaniasis (DCL), braziliensis, either concurrently or even several
mucocutaneous leishmaniasis (MCL), and years after the resolution of skin lesions. It
visceral leishmaniasis (VL). The wide spectrum may be also due to the contiguous spread of
of symptoms manifested by leishmaniasis is cutaneous leishmaniasis caused by L. tropica.
often compared to leprosy, where the localized Involvement of the mucous membranes of the
CL is similar to tuberculoid leprosy, and DCL nasal and oral cavities results in nasal stuffiness,
is similar to lepromatous leprosy. discharge, epistaxis, and destruction of the
The immune response of the host against nasal septum. This disfiguration is often called
the infection depends on Leishmania-specific espundia. Progression into the pharynx and
Th1-type CD4+ T-cells, macrophages, and larynx may threaten the airway passage, and
cytokines. However, other factors such as may lead to dysphonia, dysphagia, and even
genetics, nutritional status, and environmental aspiration pneumonia.
factors may affect the outcome of infection. Lesions usually manifest with few parasites.
Systemic Th1 response is strong in cases of
A. Cutaneous Leishmaniasis
MCL, with increased levels of peripheral
Cutaneous leishmaniasis is the most mononuclear cells in the blood.
common form of the disease, and is caused D. Visceral Leishmaniasis
by several species of Leishmania, including L.
tropica (dry or urban oriental sore), L. major Visceral leishmaniasis (or kala azar), is a
(moist or rural oriental sore), and L. mexicana disseminated parasitosis primarily caused by L.
(chiclero ulcer, usually affecting the ears). The donovani complex: L. donovani, L. chagasi, and
126 MedICal parasItology In the phIlIppInes

L. infantum. It has an incubation period of 2 to found useful. Animal inoculation using


8 months, but clinical symptoms in previously hamsters could detect low intensity of infection.
infected but asymptomatic persons may The leishmanin skin test (Montenegro
appear during immunocompromised states. skin test) can be used to identify exposure to
This manifestation of the disease stems from the parasite. It is usually positive in cases of CL
the spread of parasites into the bone marrow, and MCL, but is negative in cases of DCL and
spleen, and liver. kala azar.
In the acute phase, twice-daily fever spikes Immunologic assays such as ELISA and
(double quotidian), with accompanying chills rk39 antigen dipstick test have demonstrated
may be present, which might be mistaken for high sensitivity and specificity for VL in
malaria. During the subacute and chronic certain immunocompetent patient populations.
course, common signs and symptoms include Direct agglutination, urine antigen assays, and
fever, weakness, loss of appetite, weight loss, newer techniques such as flow cytometry and
hemorrhage, and abdominal enlargement molecular diagnostic modalities (polymerase
associated with hepatosplenomegaly. chain reaction, RFLP analysis) are also being
Phagocytosed amastigotes are present only used; the latter may be used to identify the
in small numbers in the blood. However, they species of Leishmania.
are numerous in the reticuloendothelial cells of
Treatment
the spleen, liver, lymph nodes, bone marrow,
intestinal mucosa, and other organs. In patients Primary pharmacologic treatment is
with VL, Leishmania-specific Th1 response is based on antimony compounds, notably the
usually low or absent. VL, if left untreated, has pentavalent antimonials: sodium stibogluconate
a greater than 95% mortality rate. and n-methyl-glucamine (meglumine). These
Post-kala azar dermal leishmaniasis (PKDL) drugs are still being used in areas where
is a sequela of visceral leishmaniasis, usually seen susceptibility is still good, due to its low
in endemic areas. It manifests as a cutaneous cost. However, primary treatment failure and
eruption resulting in hypopigmented macules, relapses are often observed using these drugs,
malar erythema, nodules, and ulcerations. These especially in patients with AIDS. Side effects
lesions usually manifest a few months to several such as abdominal pain, nausea, arthralgia,
years after treatment. and even fatal arrhythmias are high using
these drugs, and treatment should only be
Diagnosis
done after consultation with infectious disease
Diagnosis of active leishmaniasis is based on experts. Treatment with the antimonial drugs
the microscopic demonstration of Leishmania requires daily intramuscular or intravenous
from lesion and tissue scrapings, aspirates, or administration for up to 4 weeks, and hospital
biopsy. Giemsa and hematoxylin-eosin stains confinements are necessary.
are often used in microscopic and histologic In cases where there is treatment failure
samples, and the demonstration of amastigotes with antimonials, or in areas where resistance
confirms the diagnosis of leishmaniasis. Cultures is high, intravenous amphotericin B is the drug
are unreliable due to the difficulty of isolating of choice. Amphotericin B has a high cure rate;
the parasites, especially in old lesions. There are however, the associated side effects, as well as the
however reports of successful primary isolation cost and availability of the drug are significant
of the New World cutaneous leishmania using limiting factors. Lipid-based preparations of the
the Novy, MacNeal, and Nicolle medium drug (AmBisome) are currently being utilized as
(NNN). The Schneider’s medium was also a highly effective, better tolerated, and overall
Chapter 2: protozoan Infections 127

cost-effective drug formulation for cutaneous mostly poor and malnourished children below
and visceral leishmaniasis. 15 years old.
In India, where sodium pentavalent Leishmaniasis is primarily a disease of
antimony resistance is high, the antineoplastic poverty. It affects people living in squalid
drug miltefosine was introduced in 2002 to treat conditions, and is associated with poor housing,
VL. Miltefosine is the only oral drug currently malnutrition, a weak immune system, and
given to VL patients. lack of resources. Environmental changes such
Pentamidine is another second-line drug as deforestation, new irrigation schemes, and
for cutaneous as well as the visceral form of the urbanization are also linked to changes in the
disease. However, due to side-effects and the epidemiology of the disease. In urban areas
development of drug resistance, pentamidine where leishmaniasis occurs, there is a greater
use has been limited. For the cutaneous form of epidemic threat.
leishmaniasis, topical paromomycin has shown Visceral leishmaniasis is an important
efficacy in certain areas. opportunistic infection in AIDS patients. VL/
Combination therapy using two or more of HIV co-infection is currently a major threat in
the anti-leishmanial drugs is being studied. The the control and prevention of either disease.
presence of drug resistance especially towards Immunosuppression from HIV predisposes
the pentavalent antimonials, poor treatment to VL, while VL infection accelerates HIV
outcomes of complicated cases (such as HIV replication and progression to AIDS. VL/
coinfection), the potential for greater efficacy, HIV co-infection has been documented in
better compliance, and fewer side effects are 35 countries, with most cases coming in from
reasons why combination therapy for VL Ethiopia, southern Europe (Spain, Italy, France,
is the current consensus. Among the drug and Portugal), and Brazil.
combinations currently being used or under In the Philippines, there have been
clinical trials are: sodium stibogluconate plus imported cases of cutaneous lesions referred
paromomycin, and liposomal amphotericin B to the University of the Philippines—College
plus either miltefosine, or sodium stibogluconate. of Public Health, where amastigotes were
identified from the patients.
Epidemiology
Prevention and Control
Leishmaniasis is a global disease distributed
across 88 countries in four continents. It affects Preventive measures against leishmaniasis
more than 12 million people worldwide, and include usage of insect repellants containing
more than 350 million are at risk for the DEET and permethrin, insecticide-treated
disease. New cases of cutaneous leishmaniasis clothing, and fine-mesh bed nets. Use of fine
number between 1 to 1.5 million per year, the mesh screens and spraying of houses and
majority of which occur in Afghanistan, Brazil, buildings are also being done in certain areas.
Iran, Peru, Saudi Arabia, and Syria. American However, interval spraying predisposes to
soldiers deployed in Afghanistan and Iraq have resistance of sandflies to the insecticides, not
also demonstrated cases of CL. Mucocutaneous to mention the impact of insecticides on the
leishmaniasis occurs in Bolivia, Brazil, and environment.
Peru, while half a million new cases annually Regulation of reservoir hosts is another
of visceral leishmaniasis occur primarily in important aspect in the control and prevention
Bangladesh, Brazil, India, Nepal, and Sudan. of leishmaniasis. Insecticide-treated dog collars,
In 2009, there was a noted upsurge in VL cases mass testing of domestic dogs, and even
in Sudan compared to previous years, affecting extermination of infected dogs are current
128 MedICal parasItology In the phIlIppInes

strategies that address zoonotic transmission Markell EK, Voge M, John DT. Medical
of the disease. parasitology. 9th ed. Philadelphia: W. B.
At present, there is no commercially Saunders Company; 1992.
available form of either active or passive Nantulya VM. TrypTect CIATT a card indirect
chemoprophylaxis against leishmaniasis. agglutination trypanosomiasis test for
However, in immunocompetent individuals, diagnosis of Trypanosoma gambiense and
a form of immunity persists after resolution T. rhodesiense infections. Trans R Soc Trop
of active lesions. Certain countries, such as Med Hyg. 1997;9(1):551–3.
endemic areas in the Middle East, have been Neva FA, Brown HW. Basic clinical parasitology.
using live parasites either from infected insect 6th ed. Connecticut: Appleton & Lange;
vectors, or in recent years, from cultures, to 1994.
inoculate inconspicuous areas (such as the Roberts LS, Janovy J. Foundations of
buttocks) so as to protect themselves from parasitology. 5th ed. Dubuque: Wm. C.
disfiguring facial lesions from future infections. Brown Publishers; 1996.
Commercial vaccines are currently under Wilson WR, Sande MA. Current diagnosis
development. and treatment in infectious diseases. USA:
Lange Medical Books, McGraw-Hill; 2001.
References
p. 842–53.
Beaver PC, Jung RC, Cupp E.W. Clinical World Health Organization. WHO Fact
parasitology. 9th ed. Philadelphia: Lea & Sheet no. 116. Geneva: World Health
Febiger; 1984. Organization; 1999.
Leyritana, KT, Saniel MC, Carpo BG, Murray World Health Organization. Chagas disease:
HW. New world cutaneous leishmaniasis interruption of transmission. Wkly
in a traveler: the first documented case in Epidemiol Rec. 1998;73(1-2):1–4.
the Philippines. Acta Med Philipp. 2011; World Health Organization. Leishmaniasis:
45(3):73–6. second generation vaccines. TDR news.
Mahmoud AA. Tropical and geographical 2001;65:13.
medicine companion handbook. 2nd ed. World Health Organization. Miltefosine—1,200
Singapore: McGraw-Hill Book Co.; 1993. patients in Phase IV trial in Inidia. TDR
news. 2002;69:12.

You might also like