Anda di halaman 1dari 40

Toxoplasma gondii

280  José G. Montoya, John C. Boothroyd, and Joseph A. Kovacs

SHORT VIEW SUMMARY


Definition patients at risk for reactivation. trimethoprim-sulfamethoxazole (TMP-SMX) (IV
• Toxoplasma gondii is a ubiquitous coccidian Immunoglobulin M (IgM)-positive test results or PO; 5 mg/kg of TMP every 12 hours).
protozoan that usually causes asymptomatic should be confirmed at a reference laboratory Corticosteroids are given only for clinically
infection in humans but can cause significant (e.g., the Palo Alto Medical Foundation– significant edema or mass effect, and
disease in congenitally infected infants and Toxoplasma Serology Laboratory [PAMF-TSL]; anticonvulsants are given only after a seizure.
immunodeficient patients and occasionally in www.pamf.org/serology/). • HIV patients: Start antiretroviral therapy (ART)
immunocompetent individuals. • Maternal infection is often asymptomatic; after 2 to 3 weeks; stop anti-Toxoplasma
serology shows acute infection, and IgM- medications if the CD4 count is greater than
Epidemiology
positive test results must be confirmed at a 200 cells/mm3 for more than 6 months. High
• Toxoplasmosis is a worldwide zoonosis that
reference laboratory. Congenital infection may relapse rate occurs without ART and
can infect a wide range of animals and birds.
be asymptomatic or appear with neurologic or maintenance therapy.
• Transmission to humans is mainly by ingestion
ocular manifestations; this form is diagnosed • Acute infection in pregnant women less than
of viable tissue cysts in meat or of food or
in utero by PCR of amniotic fluid or after birth or equal to 18 weeks of gestation: Give
water contaminated with oocysts. Less
by serology or PCR. spiramycin (1 g every 8 hours; available at no
commonly, there is congenital transmission or
• Chorioretinitis may be asymptomatic or show cost through the PAMF-TSL and the U.S. Food
transplantation of an infected organ.
visual loss; ophthalmologic examination and and Drug Administration) until delivery. If
• Positive immunoglobulin G (IgG) representing
PCR of vitreous or aqueous fluid are infection in the fetus is documented or
prior infection increases with age;
performed. The Goldmann-Witmer coefficient suspected, or if at greater than 18 weeks of
seroprevalence is ≈11% in the United States
(anti-Toxoplasma IgG/total IgG in aqueous gestation, give pyrimethamine (50 mg every
and up to ≈78% in other parts of the world.
fluid divided by anti-Toxoplasma IgG/total IgG 12 hours for 2 days, then 50 mg/day) plus
• Congenital transmission occurs almost
in serum) can be helpful. sulfadiazine (initial dose 75 mg/kg, followed
exclusively when a seronegative mother
• HIV-infected patients usually present with focal by 50 mg/kg every 12 hours; maximum, 4 g/
becomes infected during pregnancy.
neurologic symptoms. Patients are usually IgG day), plus folinic acid (10 to 20 mg/day).
Retinochoroiditis can occur after congenital
positive and IgM negative, computed Before 14 to 18 weeks of gestation, give no
infection or recently acquired infection.
tomography or magnetic resonance imaging pyrimethamine or leucovorin.
Infection in human immunodeficiency virus
may show one or more contrast-enhancing • Congenitally infected infant: Give
(HIV)/acquired immunodeficiency syndrome
lesions, cerebrospinal fluid PCR is specific but pyrimethamine (1 mg/kg every 12 hours for 2
patients almost always results from
not sensitive, brain biopsy sensitivity is days, then 1 mg/kg/day for 2 or 6 months); then
reactivation of latent infection. Among organ
improved by immunoperoxidase staining, and this dose is given every Monday, Wednesday,
transplant patients, disease can result from
diagnosis is often presumptive and extends to a and Friday; plus sulfadiazine (50 mg/kg every
either newly acquired infection from the
response to empirical therapy. 12 hours) plus folinic acid (10 mg three times
transplanted organ or from reactivation of
• Immunodeficient patients present with weekly) for at least 12 months.
latent infection.
encephalopathy, seizures, pneumonia, and fever. • Chorioretinitis patients: If therapy is clinically
Microbiology Diagnosis is based on positive PCR or indicated, give pyrimethamine (100-mg
• Toxoplasma organisms are exclusively histopathology. Hematopoietic stem cell loading dose over 24 hours, then 25 to 50 mg/
intracellular. The sexual phase occurs in felines. transplantation (HSCT) requires pretransplant day) plus sulfadiazine (1 g every 6 hours) plus
Excreted oocysts require 1 to 5 days to serology; solid-organ transplantation requires leucovorin (10 to 20 mg/day) for 4 to 6 weeks.
become infectious. Tachyzoites actively pretransplant serology in the donor and recipient. TMP-SMX, one double-strength tablet every 3
replicate in essentially all cell types. Tissue days, can prevent relapse.
Therapy
cysts with intracystic bradyzoites maintain
• Immunocompetent patients: These patients Prevention and Prophylaxis
organism viability during latent infection.
usually require no therapy if asymptomatic; • Avoid undercooked meat and potentially
• Tachyzoites replicate well in tissue culture and
they may benefit from treatment if symptoms contaminated food or water; clean cat litter
are responsible for clinical manifestations
are severe or persist. daily.
during primary infection or reactivation of
• Immunocompromised patients: The therapy • Immunocompromised patients: Give TMP-SMX,
latent infection.
dosage is pyrimethamine (200-mg load, then one double-strength or single-strength tablet
• Multiple strains are identified by genotyping.
50 to 75 mg/day) plus sulfadiazine (1000 to daily. An alternative is dapsone (50 mg/day)
Strains differ in virulence, with the most
1500 mg every 6 hours) plus leucovorin (10 to plus pyrimethamine (50 mg/wk) plus
virulent strains so far reported being found in
20 mg/day). The dosage is then decreased to leucovorin (25 mg/wk). If the patient has HIV,
South America.
maintenance dosing of pyrimethamine (25 to start if the CD4 count is less than 100 to 200
Diagnosis 50 mg/day) plus sulfadiazine (500 to 1000 mg cells/mm3; discontinue if the patient is on ART
• Direct detection of the organism is by every 6 hours) plus leucovorin (10 to 20 mg/ and the CD4 count is greater than 200 cells/
polymerase chain reaction (PCR) assay, day) after 3 to 6 weeks if a clinical response mm3 for at least 3 months (primary
histopathology with immunoperoxidase occurs. Alternatives include pyrimethamine, as prophylaxis) or 6 months (secondary
staining, or, less commonly, by tissue culture or above, plus leucovorin plus either clindamycin prophylaxis). For HSCT recipients, start
mouse inoculation. (intravenous [IV] or oral [PO]; 600 mg every 6 TMP-SMX after engraftment. If the patient is a
• Serologic assays can help distinguish acute hours) or atovaquone (1500 mg every 12 solid-organ transplant recipient, start at
from chronic infection and can identify hours). Another alternative is transplantation if classified as D+ or R+.

3122
Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3122.e1
KEYWORDS
acquired immunodeficiency syndrome; antiretroviral therapy;
bradyzoite; cats; chorioretinitis; dapsone; encephalitis; lamb; oocyst;

Chapter 280  Toxoplasma gondii


pork; pregnancy; pyrimethamine; spiramycin; tachyzoite; tissue cyst;
Toxoplasma gondii; toxoplasmosis; trimethoprim-sulfamethoxazole;
zoonosis

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3123
Although Toxoplasma gondii infects a large proportion of the world’s sequence analysis indicates there is a varied amount of genetic exchange
human population, it is an uncommon cause of disease. Certain indi- within and between these clades.20 Hence, although the “three-
viduals, however, are at high risk for severe or life-threatening disease dominant-strain” paradigm is holding for humans in Europe and
because of this parasite. These include congenitally infected fetuses and North America, the situation appears much more complex in other

Chapter 280  Toxoplasma gondii


newborns and immunologically impaired individuals. Congenital regions and in other animal hosts.
toxoplasmosis is the result of maternal infection acquired during gesta-
tion, an infection that is most often clinically inapparent. In immuno- ORGANISM STAGES
deficient patients, toxoplasmosis most often occurs in persons with Oocyst
defects in T-cell–mediated immunity, such as those receiving cortico- Cats eventually shed oocysts after they ingest any of the three forms of
steroids, anti–tumor necrosis factor (TNF) therapies, certain monoclo- the parasite, at which time an enteroepithelial cycle begins. This sexual
nal antibodies, or cytotoxic drugs, and in those with hematologic form of reproduction begins when the parasites penetrate the epithelial
malignancies, organ transplants, or acquired immunodeficiency syn- cells of the small intestine and initiate development of asexual and
drome (AIDS). In the vast majority of otherwise immunocompetent sexual (gametogony) forms of the parasite. Oocyst wall formation
individuals, primary or chronic (latent) infection with T. gondii is begins around the fertilized gamete, and when still immature, oocysts
asymptomatic; after the acute infection, a small percentage suffer cho- are discharged into the intestinal lumen by rupture of intestinal epi-
rioretinitis; lymphadenitis; or, even more rarely, hepatitis, myocarditis, thelial cells.11 Unsporulated oocysts are subspherical to spherical and
and polymyositis.1 measure 10 × 12 µm in diameter (see Fig. 280-1A). Oocysts are formed
T. gondii was first observed in the North African rodent (Ctenodac- in the small intestine only in felids and are excreted in the feces for
tylus gundi) by Nicolle and Manceaux in 19082 and was recognized as periods varying from 7 to 20 days. More than 100 million oocysts may
a cause of human disease in an 11-month-old congenitally infected be shed in the feces in a single day.11 Sporulation, required for oocysts
child by Janku in 1923.3 It was reported as a cause of encephalitis by to become infectious, occurs outside the cat within 1 to 5 days, depend-
Wolf, Cowen, and Paige, who in 1939 observed it in a newborn who ing on temperature and the availability of oxygen. Sporulated oocysts
presented with seizures, intracranial calcifications, hydrocephalus, and contain two sporocysts (see Fig. 280-1A), each of which contains four
chorioretinitis.4 sporozoites. Maturation is more rapid at warm temperatures (2 to 3
Although relatively few cases of severe toxoplasmosis in adults were days at 24° C compared with 14 to 21 days at 11° C).11 Oocysts may
reported during the ensuing years, the remarkable report in 1968 by remain viable for as long as 18 months in moist soil; this results in an
Vietzke and his colleagues,5 from the National Cancer Institute of the environmental reservoir from which incidental hosts may be infected.
National Institutes of Health, highlighted T. gondii as a cause of life- Recent clues to some of the features that make oocysts so robust have
threatening infection in patients with malignancy, predominantly in come from proteomic and transcriptomic analyses.21,22 Among other
those with hematologic malignancies. Brain involvement with focal findings, these studies have revealed an abundance of small tyrosine-
areas of encephalitis was the primary finding at autopsy in these rich proteins in the oocyst wall. Cross-linking of the tyrosines in these
patients. Since that time, several hundred cases in non-AIDS immuno- proteins could confer a natural “sun-screen” for the sporozoites within
deficient patients have been recorded in the literature.6 In 1983, the first because they are strong absorbers of ultraviolet light.
report of toxoplasmosis in AIDS patients appeared.7 Toxoplasmic
encephalitis (TE) subsequently was recognized as the major cause of Tachyzoite
space-occupying lesions in the brains of these patients, almost all of The tachyzoite form (see Fig. 280-1B) is oval to crescentic and mea-
whom had serologic evidence of prior exposure to the parasite.7 Despite sures 2 to 3 µm wide and 5 to 7 µm long; it requires an intracellular
the significant advances that have been achieved in the recent past, habitat to multiply despite having all of the usual eukaryotic machinery
major challenges remain in the areas of prevention and management necessary for reproduction. Tachyzoites are seen in both primary and
of the acute infection in pregnancy, the fetus, and the newborn,8 and reactivated infection; their presence is the hallmark of active infection.
in the understanding and treatment of toxoplasmic chorioretinitis9 and They reside and multiply within vacuoles in their host’s cells, can infect
infection in immunocompromised individuals.1,6,10 virtually all phagocytic and nonphagocytic cell types,11 and multiply
approximately every 6 to 8 hours to form rosettes.23 Continuous mul-
ETIOLOGY tiplication leads to cell disruption and release of organisms that go on
T. gondii is a coccidian parasite of felids with humans and other warm- to invade nearby cells or are transported to other areas of the body by
blooded animals serving as intermediate hosts. It belongs to the sub- blood and lymph.24 Tachyzoites appear to actively and rapidly migrate
phylum Apicomplexa, class Sporozoa, and exists in nature in many across epithelial cells and may traffic to distant sites while extracellu-
forms: macrogametes and microgametes, the oocyst (which releases lar.25 Recent evidence suggests they might also use the infected host
sporozoites), the tissue cyst (which contains and may release bradyzo- cell as a “Trojan horse” to traffic and gain access to tissues that might
ites), and the tachyzoite (Fig. 280-1).11 not otherwise be easily accessed.26
Population genetic analysis has demonstrated that, at least within At the anterior end of the tachyzoite, there is a cone-shaped struc-
Europe and North America, most organisms isolated from both ture termed the conoid. It is protruded during the parasite’s entry into
domesticated animals and humans can be grouped into one of three host cells. Rhoptries, numbering 4 to 12, are club-shaped organelles
clonal genotypes—types I to III—that may identify clinically relevant that terminate within the conoid. The rhoptries, together with sur-
biologic differences.12 Clear differences have been observed in the fre- rounding small, rod-shaped organelles (micronemes), have important
quency of parasite genotypes when T. gondii isolates from animals were secretory functions for parasitic invasion. Dense granules are organelles
compared with those of humans. Type III strains are common in distributed throughout the cytoplasm. Their contents are released into
animals but observed significantly less often in cases of human toxo- a vacuole, termed the parasitophorous vacuole, that is formed around
plasmosis; most cases in humans in Europe and North America are the parasite during entry into the cell and also into the external envi-
caused by type II strains. Type II strains are significantly more often ronment as excreted-secreted antigens.11
associated with reactivation of chronic infections and accounted for The rhoptries and micronemes produce a collection of proteins
65% of strains isolated from AIDS patients.13 Both type I and type II that are crucial for the invasion process.27 These appear to mediate
strains have been associated with human congenital toxoplasmosis.13-15 the attachment to the host cell, including the moving junction, a
To date, types II and III have not been detected in immunocompetent ringlike point of contact between the parasite and host cell surface
individuals with severe ocular disease.16 Atypical and recombinant that migrates down the length of the parasite during invasion. The
strains have been identified with increasing frequency in regions other rhoptries also inject proteins into the host cell that are critical in
than the United States and Europe and from animals other than manipulating the host cell, presumably to the advantage of the para-
humans and domestic animals; some of these strains appear to be site.28 These rhoptry proteins are very different (polymorphic) between
associated with more severe disease, suggesting greater virulence, even different strains of T. gondii and appear responsible for many of the
in immunocompetent individuals.17,18 The most exhaustive studies differences in virulence seen for types I, II and III in mice, as described
have now identified six major population clades,19 although detailed earlier.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Part III  Infectious Diseases and Their Etiologic Agents 3124

A B

C D
FIGURE 280-1  The three forms of Toxoplasma gondii observed in nature. A, Oocysts. Unsporulated oocyst (top left). Sporulated oocyst with
two sporocysts (top right). Four sporozoites (arrows) are visible in one of the sporocysts. Transmission electron micrograph of a sporulated oocyst (bottom).
Note the thin oocyst wall (large arrow), two sporocysts (arrowheads), and sporozoites, one of which is cut longitudinally. B, Giemsa stain demonstrating
two rosettes of intracellular tachyzoites in a mouse bone marrow macrophage. C, Giemsa-stained smear of mouse peritoneal fluid demonstrating
the tachyzoite form. D, Hematoxylin and eosin stain of the cyst form in brain. (A courtesy Dr. J.P. Dubey, U.S. Department of Agriculture. Beltsville, MD.
B to D courtesy Dr. Jack S. Remington, Stanford University and Palo Alto Medical Foundation.)

Tachyzoites cannot survive desiccation, freezing and thawing, or and synchronously, forming rosettes and lysing the cell, whereas the
extended exposure to gastric digestive juices.24 They are propagated more slowly replicating bradyzoites form tissue cysts. Molecules are
in the laboratory in the peritoneum of mice and in cultured cells. expressed in a stage-specific manner and are responsible for certain of
Tachyzoites can be visualized in sections stained with hematoxylin and the phenotypic differences between tachyzoites and bradyzoites.
eosin but are better visualized with Wright-Giemsa and immunoper- Interferon-γ (IFN-γ), nitric oxide (NO), heat shock proteins, and pH
oxidase stains.29 and temperature manipulations can trigger conversion of tachyzoites
A fourth organelle that is restricted to Toxoplasma and its Apicom- to bradyzoites in vitro and perhaps in vivo as well.11
plexa cousins is the apicoplast.30 This is akin to chloroplasts of plants, Tissue cysts grow and remain within the host cell cytoplasm as the
with a similar evolutionary origin involving endosymbiotic algae, but intracystic form wherein the bradyzoites continue to divide. Tissue
photosynthetic functions have been completely lost. It has its own cysts vary in size from younger ones that contain only a few brady­zoites
DNA, RNA, and protein translation, the latter of which is prokaryotic to older tissue cysts that may contain several thousand brady­zoites and
in nature, making for a very attractive target for drug therapies. Indeed, may reach more than 100 µm in size (see Fig. 280-1D). They appear
one of the currently used drugs for treatment of human infection, spherical in the brain and conform to the shape of muscle fibers in heart
clindamycin, targets the ribosomes of the apicoplast.31 A major role of and skeletal muscles. The central nervous system (CNS); eye; and skel-
this organelle is fatty acid biosynthesis.30 Recent work with Plasmo- etal, smooth, and heart muscles appear to be the most common sites of
dium, which also has this organelle, has demonstrated that the apico- latent infection.33 Because of their persistence in tissues, demonstration
plast is crucial to the synthesis of isoprenoids,32 making further work of tissue cysts in histologic sections does not necessarily mean that the
on attacking this Achilles heel an attractive prospect. infection was recently acquired or that it is clinically relevant. Tissue
cysts stain well with periodic acid–Schiff, Wright-Giemsa, Gomori
Tissue Cyst methenamine silver, and immunoperoxidase stains. Tissue cysts in
Once the tachyzoite has invaded the target cell, it can undergo stage meat are rendered nonviable by γ-irradiation (0.4 kGy),34 heating meat
conversion into the bradyzoite form.11 Tachyzoites and bradyzoites are throughout to 67° C, or freezing to −20° C for 24 hours and then
structurally and phenotypically different. Tachyzoites multiply rapidly thawing,35,36 but not by gentle heating in a microwave.37

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3125
Although the tachyzoite form appears to be indiscriminate in the place in the small bowel of members of the cat family, cats play a sig-
type of host cell parasitized, it has been suggested that, in brain tissue, nificant role as powerful amplifiers of the infection in nature (see
there is a predilection for tissue cyst formation to occur predominantly “Oocyst”).11 Epidemiologic surveys have revealed that in most areas of
within neurons.38,39 However, it has been shown that tissue cysts can the world, the presence of cats is of primary importance for the trans-

Chapter 280  Toxoplasma gondii


form within astrocytes cultured in vitro.40 In an electron microscopic mission of the parasite. Excretion of oocysts has been reported to occur
study of the pathologic changes in brains of infected mice, tissue cysts in approximately 1% of cats in diverse areas of the world.45 Wild felines,
were observed to remain intracellular throughout the period of study and especially bobcats in the United States, may be among the most
(22 months).38 There is compelling evidence to suggest that bradyzoites important sources of oocysts.46
can exit from intact tissue cysts and invade contiguous cells, where they Although ingestion of raw or undercooked meat that contains
convert to the tachyzoite form.41 This is the likely explanation for the viable T. gondii tissue cysts will result in infection, the relative fre-
appearance of “daughter” cysts or clumps of cysts in the brain. Recent quency with which this occurs in relation to the frequency of infection
evidence suggests that neurons are capable of destroying the invaded caused by ingestion of oocysts is unclear. For instance, in countries
parasite and/or that rhoptry proteins are injected into cells they do not such as France, where eating undercooked meat is common and the
infect.42 These results have implications for how the parasite comman- prevalence of the infection is high, meat may be an important cause of
deers host functions. the infection. (It was in Paris that the meat-to-human hypothesis of
spread of T. gondii was proved.47) In contrast are countries such as
TRANSMISSION AND those in Central and South America, where the prevalence of the infec-
EPIDEMIOLOGY tion in humans is high but the ingestion of undercooked meat is
T. gondii infection is a worldwide zoonosis. The organism infects her- uncommon. In these areas, oocysts may be the more important source
bivorous, omnivorous, and carnivorous animals, including birds.43 of human infection.48 Until a newly described method for distinguish-
Infection in humans most commonly occurs through the ingestion of ing bradyzoite- versus oocyst-initiated infection is validated in these
raw or undercooked meat that contains tissue cysts, through the inges- regions,49 their respective contributions to human infection will largely
tion of water or food contaminated with oocysts, or congenitally remain a matter of speculation.
through transplacental transmission from a mother who acquired her Ingestion of tissue cysts in infected meat (primarily pork and lamb)
infection during gestation (Fig. 280-2). Less common are transmission is a major source of the infection in humans in the United States.50,51
by transplantation of an infected organ or transfusion of contaminated T. gondii infection is common in many animals used for food, espe-
blood cells. Transmission has also occurred by accidental sticks with cially sheep and pigs, with a lower prevalence in cattle, horses, and
contaminated needles44 or through exposing open lesions or mucosal water buffaloes. Infection resulting in transmission to humans has also
surfaces to the parasite.45 Because the sexual cycle of the parasite takes been documented in wild game animals.52 Organisms may survive in

Raw milk

Meat, other
tissue
Untreated Fruits and
water vegetables

Vertical
transmission
Soil Raw shellfish

Oocyst
Tissue cysts

Cat feces

Oocyst

Grazing

Oocyst Tissue cysts


Carnivorism

FIGURE 280-2  Transmission and life cycle of Toxoplasma gondii.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3126
tissue cysts in these animals for years and can be found in nearly all and at high elevations. Slaughterhouse workers may have an increased
edible portions of an animal.53 A seminal study on the prevalence of risk for infection. The prevalence of antibody titers to T. gondii varies
T. gondii in samples of meat used for human consumption (obtained considerably among different geographic areas and also among indi-
from grocery stores) was performed in the United States in the 1960s.54 viduals within a given population. These differences depend on a
Part III  Infectious Diseases and Their Etiologic Agents

The parasite was isolated from 32% of pork chops and 4% of lamb variety of factors, including culinary habits and cleanliness of sur-
chops; there were no isolations from beef, and indeed, cattle appear roundings. A decrease in antibody prevalence over the past few decades
to be generally not an important intermediate host for this parasite.55 has been observed in many countries. In the United States, the sero­
A recent polymerase chain reaction (PCR)-based study in England prevalence in U.S. military recruits decreased by one third between
found 33% (19/57) of pork and 67% (6/9) of lamb samples positive for 1965 and 1989; the crude seropositivity rate among recruits from 49
T. gondii DNA.56 states was 9.5% in 1989 compared with 14.4% in 1965.77 As another
Serologic surveys conducted in the past 20 years in the United example, in the 1970s, 24% of women in the childbearing age group in
States indicate that the prevalence of T. gondii in pigs is declining, with Palo Alto, California were seropositive, whereas the rate in 2008 was
an overall prevalence of 2.6% in a recent National Animal Health 10%. Seroprevalence rates in the United States among such women
Monitoring Survey.57 This reduction in T. gondii prevalence has been range from 3% to greater than 35%, whereas rates greater than 50% are
attributed to changing management practices and consolidation of pig present in women of childbearing age in much of Western Europe,
production into large-scale operations. However, there are still many Africa, and South and Central America.78 The recent (1999 to 2004)
isolated small swine farms, including those that raise organic pigs, and overall age-adjusted seroprevalence of T. gondii infection in the United
the prevalence of T. gondii in these animals can be greater than 90%.58 States, based on a study of 15,960 persons aged 6 to 49, was reported
Of note, meat for human consumption is not routinely inspected for to be 10.8%, with a seroprevalence among women aged 15 to 44 years
T. gondii infection in the United States or elsewhere in the world.59 of 11%.79 This study found an approximately 25% to 40% decline in
Seroprevalence of T. gondii infection in a study of lambs in the mid- seroprevalence compared with a similar survey a decade earlier.
Atlantic region was recently reported to be 27%.60 Although T. gondii Although the prevalence of the infection appears to be declining in
infection of sheep is widely prevalent, the public health importance of certain areas of the world, such as Europe and the United States, this
this is unclear because, in the United States, meat from adult sheep is has not been the case or it has been documented to have increased in
not usually used for human consumption.45 Reports of suspect trans- other geographic locales.78
mission by unpasteurized goat’s milk have appeared.61,62 In addition to T. gondii may survive in citrated blood at 4° C for as long as 50 days,
differences in how meat is cooked, the tendency for beef to harbor few, and infection has been transmitted through transfusion of whole blood
if any, cysts compared with lamb and pork may partly explain the dif- or white blood cells. Leukocyte transfusions may pose a special risk.80
ferences in seroprevalence in the United States versus Europe; beef The transmission of infection by organ transplantation has been docu-
accounts for a much greater fraction of meat consumed in the United mented and may result from the transplantation of an organ (e.g., heart)
States compared with Europe, where lamb and pork are more popular. from a seropositive donor to a seronegative recipient.81 In bone marrow
T. gondii infection is prevalent in game animals, especially black transplant recipients, toxoplasmosis almost always is a result of recru-
bears (80% infected) and white-tailed deer, as well as in raccoons (60% descence of a latent infection rather than from the transplant.82,83
infected).53 Infection in raccoons and bears is a good indicator of the The incidence of TE among HIV-infected individuals directly cor-
prevalence of T. gondii in the environment because these animals are relates with the prevalence of T. gondii antibodies among the general
scavengers. Thus, wild animal meat can serve as a source of the infec- HIV-infected population, the degree of immunosuppression (best
tion for hunters and their families, especially when care is not taken measured by the CD4 cell count),84 the use of effective prophylactic
while eviscerating and handling the game or when meat and other treatment regimens against development of TE, and the immunologic
organs from these animals is served undercooked or uncooked.53 response to antiretroviral therapy (ART).85 AIDS-associated TE and
Although T. gondii tissue cysts may be found in edible tissues of toxoplasmosis involving other organs are almost always due to reacti-
chickens,63 poultry products are probably not important in the trans- vation of a chronic (latent) infection that results from the progressive
mission of T. gondii to humans because they are usually frozen for immune dysfunction that develops in these patients.86 It is estimated
storage and thoroughly cooked to avoid diseases that could be caused that 20% to 47% of AIDS patients who are infected with T. gondii but
by contamination by other organisms.45 The parasite has been isolated are not taking anti-Toxoplasma prophylaxis or antiretroviral drugs will
from chicken eggs.64 ultimately develop TE.84,86 This makes TE a major concern in areas
The ingestion of vegetables and other food products contaminated where the use of antiretrovirals is still a treatment relatively few HIV-
with oocysts probably accounts for infection in seropositive vegetari- positive individuals receive.
ans. Although isolation of tachyzoites from secretions of people with In recent years a substantial decline in the incidence of TE87 and
the acute infection has been claimed, human-to-human transmission toxoplasmosis-associated deaths88 has been seen in HIV-infected
of infection by this route has not been established. Outbreaks within patients who adhere to effective anti-Toxoplasma prophylactic regi-
families and other groups are common,65-67 but there is no evidence mens and to ART.
of natural human-to-human transmission other than from mother In the United States, T. gondii seropositivity among HIV-infected
to fetus. patients varies from 10% to 45%86 and directly correlates with the
Several epidemiologic studies have identified water as a potential seropositivity in the general non–HIV-infected population. In con-
source for T. gondii infection both in humans and animals.65,68-71 In trast, the seroprevalence is approximately 50% to 78% in certain areas
vitro studies have demonstrated that oocysts can sporulate in seawater of Western Europe and Africa.89,90 In a study in France, 1215 (72.2%)
within 1 to 3 days, can survive in seawater for up to 6 months, and can of 1683 HIV-infected patients had serologic evidence of exposure to T.
survive in water treated with sodium hypochlorite or ozone, but not gondii.91 During the study period (1988 to 1995), the overall incidence
ultraviolet radiation.72-74 Population mapping studies of acutely infected of toxoplasmosis in this population was estimated to be 1.53 per 100
individuals as well as case-control studies linked drinking unfiltered patient-years, with an increase from 0.68 per 100 patient-years in 1988
water (presumably contaminated with oocysts) to an outbreak of toxo- to 2.1 per 100 patient-years in 1992, and a subsequent decline to 0.19
plasmosis in a municipality in the Western Canadian province of per 100 patient-years in 1995 that was likely related to the widespread
British Columbia65 and to high endemic rates of toxoplasmosis in Rio use of anti-Toxoplasma prophylaxis. Toxoplasmosis is rare in the HIV-
de Janeiro State, Brazil.69 In another Brazilian outbreak, T. gondii infected pediatric population: 0.06 cases per 100 patient-years were
organisms were detected in water by a variety of methods from an reported among more than 3000 patients participating in clinical trials
implicated reservoir.75 Coastal freshwater runoff was observed to be a in the pre-ART era but during a time when Pneumocystis carinii pneu-
risk factor for T. gondii infection among southern sea otters along the monia (PCP) prophylaxis was recommended.92
California coast.76 Of interest is the low reported incidence of TE in Africa, despite T.
In humans, the incidence of T. gondii antibodies increases with gondii seroprevalence rates of 32% to 78%. Lack of autopsy data and a
increasing age; the incidence does not vary significantly between sexes. lack of neuroimaging studies likely contribute to the low reported
The incidence tends to be less in cold regions, in hot and arid areas, incidence. It has also been suggested that because of poor access to

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3127
medical care, many HIV-infected patients in Africa succumb to infec- proliferation of tachyzoites and tissue destruction. In individuals with
tion with organisms such as Mycobacterium tuberculosis before they deficient cell-mediated immunity, rapid, uncontrolled proliferation of
develop the opportunistic infections associated with the advanced T. gondii results in progressively enlarging necrotic lesions. It has been
stage of HIV infection, including toxoplasmosis. However, in one postulated that damage to any organ in these patients, including the

Chapter 280  Toxoplasma gondii


autopsy series, from the Ivory Coast, of 175 patients with AIDS- brain, eye, heart, lung, skeletal muscle, gastrointestinal tract, and pan-
defining abnormalities, the prevalence of TE was 21%.93 creas, can result directly from tissue cyst disruption in the parenchyma
T. gondii infection may be acquired after the acquisition of HIV of the organ itself or from tissue cyst disruption elsewhere in the body,
infection. Seroconversion rates between 2% and 5.5% have been followed by subsequent spread to that organ.106 Hematogenous spread
reported in patients followed for periods up to 28 months.94 is supported by the observation of the development of simultaneous
Even before the emergence of AIDS, TE had been recognized as a lesions in the brain and the presence of parasitemia in 14% to 38% of
cause of incapacitating disease and death among HIV-negative immu- AIDS patients with TE.107,108
nosuppressed patients,6,95 especially in those whose underlying disease Infection with T. gondii induces both humoral and cell-mediated
or therapy caused a deficiency in cell-mediated immunity. Patients immune responses. A well-orchestrated and effective systemic immune
with hematologic malignancies, especially those with Hodgkin’s response, combining both innate and adaptive mechanisms, is respon-
disease, are at a particularly higher risk to develop recrudescence of sible for the early disappearance of T. gondii from peripheral blood
the infection. Among organ transplantation patients, those with heart, during the acute infection and limits the parasite burden in other
lung, kidney, and bone marrow transplants develop toxoplasmosis at a organs. Immunity in the immunocompetent host is lifelong. Exoge-
higher rate. nous reinfection, which has been demonstrated in laboratory animals,
likely also occurs in humans but does not appear to result in clinically
PATHOGENESIS AND IMMUNITY apparent disease, although, in one case report, a chronically infected
T. gondii multiplies intracellularly at the site of invasion (the gastroin- pregnant woman was infected with a highly virulent strain that resulted
testinal tract is the major route for and the initial site of infection in in infection of the fetus.109
nature); bradyzoites released from tissue cysts or sporozoites released Because T. gondii is a natural parasite of rodents, inbred mice have
from oocysts penetrate, differentiate to tachyzoites, and then rapidly been used extensively as an animal model for studies of both immunity
multiply within intestinal epithelial cells. Organisms may spread first and immunopathology in this protozoan infection and have yielded a
to the mesenteric lymph nodes and then to distant organs by invasion remarkably detailed picture of its host interaction.110-112 When tachyzo-
of lymphatics and blood. T. gondii tachyzoites infect virtually all cell ites invade, they inject the contents of their rhoptries into the host cell
types, and cell invasion occurs as an active process. Survival of tachyzo- cytosol. This delivers not only some of the machinery needed for inva-
ites is due to the formation of a parasitophorous vacuole that lacks host sion (contained within the rhoptry necks and known as RON proteins)
proteins necessary for fusion with lysosomes,96 and consequently acidi- but also a collection of “effectors” that intercept or co-opt host immune
fication does not occur. Active invasion of macrophages by tachyzoites pathways, presumably to the parasite’s advantage. These effectors
does not trigger oxidative killing mechanisms. With the appearance of include the rhoptry protein, ROP16, which functions as a mimic of
humoral and cellular immunity, only those parasites protected by an host Janus kinases (JAKs), phosphorylating critical tyrosines on host
intracellular habitat or within tissue cysts survive. An effective immune STATs (signal transducers and activators of transcription). Depending
response significantly reduces the number of tachyzoites in all tissues, on the particular flavor of ROP16 that a given strain carries, the host
and after the initial acute stages, tachyzoites are rarely demonstrable immune response can be driven in differing directions, either more or
histologically in tissues of infected immunocompetent humans. less inflammatory, and this can have a profound effect on the host’s
Tachyzoites are killed by reactive oxygen intermediates,97 acidification,98 response and ultimate outcome of the infection. In the case of another
osmotic fluctuations, reactive nitrogen intermediates,99 intracellular pair of injected ROPs, ROP5 and ROP18, the target is murine IRGs, a
tryptophan depletion,100 and specific antibody combined with comple- key part of a mouse cell’s defense machinery.113 Normally, IRGs attack
ment.101 In rodents, two classes of immune-stimulated guanosine tri- the membrane of the vacuole in which a pathogen resides, disrupting
phosphate (GTP)ases play a crucial role in destruction of tachyzoites it and leading to death of the organisms within. ROP5 and ROP18,
within the parasitophorous vacuole: the p47 immunity-related GTPases however, collaborate to phosphorylate and thereby inactivate IRGs
(IRGs)102 and the larger GTP-binding proteins (GBPs).103 although, again, the effectiveness of this depends on the specific alleles
Tissue cyst formation takes place in multiple organs and tissues of ROP5 and ROP18 that a given strain of Toxoplasma carries. Lastly,
during the first week of infection. Despite the ability to isolate T. gondii dense granules can also introduce polymorphic effectors into the host
from normal brains of chronically infected humans, the tissue cyst cell; one such effector, GRA15, has been shown to be crucial to the
form is rarely observed in histologic preparations; it has been isolated activation of one of the most central transcription factors in mam-
from both brain and skeletal muscle in 10% of 52 T. gondii–seropositive malian immune response, nuclear factor kappa B.114
patients who, at autopsy, had no clinical or pathologic evidence of the It is important to recognize that these findings do not necessarily
infection.33 The tissue cyst form is responsible for residual (chronic or represent the mechanisms underlying the immune response to T.
latent) infection and persists primarily in the brain, skeletal and heart gondii in humans. For example, although the effect of ROP16 on STATs
muscle, and the eye.33,104 may well have a parallel in human cells, IRGs are not part of the human
In immunocompetent individuals, the initial infection and the immune response, and so ROP5 and ROP18, if they are impacting the
resultant seeding of different organs leads to a chronic or latent infec- outcome of human infection, must be doing so by acting on other
tion with little, if any, clinical significance. This chronic stage of the targets, such as activating transcription factor-6β.115 Similarly, Toll-like
infection corresponds to the asymptomatic persistence of the tissue receptors TLR11 and TLR12 play a major role in the induction of
cyst form in multiple tissues. It is believed that periodically, bradyzoites interleukin-12 (IL-12) and host resistance in the mouse, but neither
are released from tissue cysts or that cysts “rupture”; cyst disruption in receptor exists in humans, indicating that innate recognition of the
this setting appears to be a clinically silent process effectively contained parasite in humans must involve distinct mechanisms. This might
by the immune system and in the CNS likely results in small inflam- involve another branch of the innate immune system, NLRs (nucleo-
matory nodules, with a limited degree of neuronal cell death and tide oligomerization domain [NOD]-like receptors), that detect molec-
architectural damage.41 However, several investigators have suggested ular signatures or patterns specific to various pathogens. Recent work
that chronic infection may not be completely asymptomatic and may has suggested that susceptibility to Toxoplasma infection in humans is
result in important behavioral changes and neuropsychiatric disor- associated with a polymorphism in a human NLR known as NALP1
ders,105 although definitive studies to support such associations have (NACHT-LRR-PYD domains–containing protein 1).116 The overall
not yet been reported. question of how Toxoplasma is sensed by the innate arm of the human
Toxoplasmosis in severely immunodeficient individuals may be immune system and how different strains of the parasite do this with
caused by primary infection or be the result of recrudescence of a latent differing degrees of success is just beginning to be explored.
infection. It is widely held that reactivation is the result of disruption T cells, macrophages, and type 1 cytokines (IFN-γ, IL-12) are
of the tissue cyst form, followed by differentiation to and uncontrolled crucial for control of T. gondii infection. Adoptive transfer and

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3128
depletion experiments in murine models not only proved that T cells impaired as a result of genetic disruptions of the IFN-γ or IFN-1 genes
are essential for control of T. gondii infection but also demonstrated an succumb to the acute infection.145,146 Similar enhanced susceptibility
interplay between CD4+ and CD8+ T cells in both the induction of was observed in mice treated with the reactive nitrogen intermediate
resistance and the maintenance of latency. Expansion of both natural inhibitor aminoguanidine147 and in nitric oxide synthase–deficient
Part III  Infectious Diseases and Their Etiologic Agents

killer (NK) and γδ T cells early in infection provides innate resistance mice.148 The protective role of NO appears to be tissue-specific rather
while the adaptive response mediated through αβ CD4 and CD8 T than systemic.148 Because control of the acute infection in vivo was
cells develops. These different subsets of T cells and NK cells are likely unaffected by nitric oxide synthase deficiency, the major role of reac-
to protect the host by secreting cytokines, such as IFN-γ, IL-2, and tive nitrogen intermediates appears to be to maintain control of estab-
TNF-α, and apparently not by lysing T. gondii–infected cells.117-120 Den- lished infections in this mouse model.148
dritic cells and inflammatory monocytes also play an important role The IFN-γ–inducible p47 GTPases IRGM3 (IGTP) and IRGM1
in control of acute infection and the early production of IL-12.120,121 (LRG47) have been shown to be required for host control of T. gondii
NK-cell–derived IFN-γ appears critical to the differentiation of IL-12– infection in the mouse,149 and recent studies have linked IRGM3 with
producing dendritic cells.120 Early studies suggested that neutrophils the autophagic destruction of Toxoplasma-containing vacuoles in
were an important component of the early response, but more recent IFN-γ–activated macrophages.150,151
studies suggest that they are not and may, in fact, contribute to the Immunoglobulin G (IgG), IgM, IgA, and IgE antibodies are pro-
pathology.121 duced in response to the infection. Extracellular tachyzoites are lysed
The co-stimulatory molecules CD28 and CD40 ligand are pivotal by specific antibody when combined with complement. In mice,
for the regulation of IL-12 and IFN-γ production in response to the humoral immunity results in limited protection against less virulent
parasite.122 T. gondii infection of antigen-presenting cells, such as den- strains of T. gondii but not against virulent strains.152
dritic cells and macrophages, causes upregulation of the counter- Both astrocytes and microglia likely play important roles in the
receptors for CD28 and CD40L, CD80/CD86, and CD40.122 Binding of immune response against T. gondii within the CNS. In the early stages
CD80/CD86 to CD28 enhances production of IFN-γ by CD4+ T cells. of TE in both humans and mice, there is a remarkable and widespread
In addition, binding of CD40L to CD40 triggers IL-12 secretion, which astrocytosis restricted to areas in which the parasite is detected.125
in turn enhances production of IFN-γ. The relevance of CD40L in the Whereas T. gondii can invade, survive, and multiply within astrocytes,
immune response to T. gondii is supported by reports of TE and dis- they are killed by activated microglia.153
seminated toxoplasmosis in children with congenital defects in CD40L
signaling (hyper-IgM syndrome).123 Moreover, recent studies have GENETIC SUSCEPTIBILITY
demonstrated that expression of CD40L is defective on CD4+ T cells The observations in mice that genetic factors in the host contribute to
from HIV-infected patients.124 This deficiency may play a role in defec- the development and severity of TE154-156 and the fact that not all HIV-
tive IL-12/IFN-γ production associated with HIV infection. infected patients with positive T. gondii serologic findings develop TE
Cytokines play a critical role in defense against the infection and suggested the possibility that genetic factors may also play a role in the
are important in the pathogenesis of toxoplasmosis and TE.125 IL-12 predisposition of AIDS patients for this disease.157 The major histo-
enhances survival of T-cell–deficient mice during T. gondii infection, compatibility complex (MHC) class II gene DQ3 (HLA-DQ3) has been
by stimulating the production of IFN-γ by NK cells,126 and is thought significantly associated with the development of TE in North American
to also regulate the expression of the latter cytokine by T cells in white AIDS patients, whereas HLA-DQ1 was marginally protective.157
immunocompetent mice.127 IFN-γ has been shown to play a significant HLA-DQ3 was also significantly associated with the development of
role in the prevention or development of TE in mice.128 The administra- hydrocephalus in children with congenital toxoplasmosis.158 In the
tion to chronically infected mice of a monoclonal antibody against latter study, a mouse model transgenic for human MHC class II found
IFN-γ resulted in a dramatic worsening in the degree of encephalitis.129 higher organism burden with the HLA-DQ3 than HLA-DQ1 genotype.
In mice with active TE, treatment with IFN-γ significantly reduced the In a South American white population, a study using a higher resolu-
inflammatory response and numbers of tachyzoites.130 tion typing method identified HLA-DQB*0402 and HLA-DRB1*08
Differences in IL-12 levels elicited during infection by different genes, which were in linkage disequilibrium, as risk factors for TE,
strains of the parasite may be responsible for some of the strain-specific whereas alleles of HLA-DQB3 were not.159 Thus, further studies are
differences in virulence in mice.131 These differences appear to be needed to better define the contribution of various HLA alleles to
related to the activation (phosphorylation) of the transcription factor susceptibility to TE. In single studies, polymorphisms in other genes,
STAT3, which in turn is dependent on the particular allele of ROP16 including those for IL-10, TLR9, NLR family, pyrin domain–containing
injected by a given strain, as detailed earlier.132 protein 1 (NALP1, also known as NLRP1), and purinergic receptor
TNF-α is another cytokine pivotal for control of T. gondii infection. P2X(7) (P2RX7) have been associated with congenital toxoplasmosis
TNF-α is required for triggering of IFN-γ–mediated activation of mac- or retinochoroiditis.116,160-162
rophages for T. gondii killing activity133 and for nitric oxide (NO, an
inhibitor of T. gondii replication) production by macrophages.134 The PATHOLOGY
administration of TNF-α neutralizing antibody to infected mice Our knowledge of the pathology of infection in humans has come
caused the death of the mice and an increase in the number of T. gondii largely from autopsy studies in severely infected infants and immuno-
tissue cysts in the brains of survivors.135 deficient patients. Data from immunocompetent adults are limited
IL-10 has been shown to deactivate macrophages and result in almost entirely to results obtained from lymph node biopsy speci-
reduced in vitro killing of T. gondii. IL-4 and IL-6, which are usually mens163 and occasionally from myocardial or skeletal muscle tissue
considered downregulatory cytokines, have been shown to be impor- specimens.164 In addition to the direct demonstration of parasites in
tant in resistance against TE in the murine model.136,137 IL-7 has also tissue and associated pathology, recent studies have revealed that the
been shown to have a protective role against T. gondii in mice.138 impact of the parasite may extend to many more cells than are actively
During the early stages of the infection, IL-12, IL-1, and TNF act in infected, that is, mouse studies have shown evidence of injected rhoptry
concert with IL-15 to stimulate NK cells to produce IFN-γ.139 proteins in upward of 50-fold more neurons in a chronically infected
IL-17140 and IL-23141,142 have also been implicated in the generation brain than actually harbor parasites at that time.42 The impact of these
of a potent immune response but are not thought be essential for host “injected-uninfected” cells on pathogenesis and other interactions with
resistance. the host has yet to be determined.
Several hypotheses have been proposed to explain the role of IFN-γ
in host resistance to T. gondii. Involvement of reactive nitrogen inter- Lymph Node
mediates (including NO) is suggested by the observation that l-NG- The histopathologic changes in toxoplasmic lymphadenitis (TL) in
monomethyl-l-arginine acetate (l-NMMA), a competitive analogue of immunocompetent individuals are frequently distinctive and often
l-arginine, simultaneously inhibits NO synthesis and intracellular diagnostic (Fig. 280-3A).163 There is a typical triad of findings: a reac-
tachyzoite killing by cytokine-activated peritoneal macrophages and tive follicular hyperplasia, irregular clusters of epithelioid histiocytes
microglial cells.99,143,144 In addition, mice in which NO synthesis is encroaching on and blurring the margins of the germinal centers, and

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3129

Chapter 280  Toxoplasma gondii


A B

C D
FIGURE 280-3  Histologic features of Toxoplasma gondii in humans. A, Hematoxylin and eosin (H&E) stain of a lymph node biopsy specimen
from an immunocompetent patient with toxoplasmic lymphadenitis. B, Positive immunoperoxidase stain of a brain biopsy specimen in a patient with
acquired immunodeficiency syndrome and toxoplasmic encephalitis. C, H&E stain of a right ventricle endomyocardial biopsy specimen from a patient
with toxoplasmic myocarditis. Organisms are seen within myocytes (see text under “Clinical Manifestations”). D, H&E stain of a right quadriceps muscle
biopsy specimen depicting tissue cyst from the same patient as shown in C. She also developed toxoplasmic polymyositis. (A courtesy Dr. Henry Masur,
Critical Care Medicine Department, National Institutes of Health, Bethesda, MD. B to D courtesy Dr. Jack S. Remington, Stanford University and Palo Alto
Medical Foundation.)

focal distention of sinuses with monocytoid cells (see Fig. 280-3A).165 peripheral zone contains T. gondii tissue cysts.171 In the areas around
Langerhans giant cells, granulomas, microabscesses, and foci of necro- the abscesses, edema, vasculitis, hemorrhage, and cerebral infarction
sis are not typically seen. Rarely, tachyzoites or tissue cysts are demon- secondary to vascular involvement may also be present.172 Important
strable. T. gondii DNA has infrequently been amplified from lymph associated features in TE are the presence of arteritis, perivascular
node tissue.166 cuffing, and astrocytosis. Because these findings may also be present
in patients with viral encephalitis, immunoperoxidase staining is
Central Nervous System important for differentiating these pathologic processes. Widespread,
Damage to the CNS by T. gondii is characterized by multiple foci of poorly demarcated, and confluent areas of necrosis with minimal
enlarging necrosis and microglial nodules.167 Necrosis is the most inflammatory response are seen in some patients.172 Identification of
prominent feature of the disease because of vascular involvement by tachyzoites is pathognomonic of active infection, but their visualiza-
the lesions. In cases of congenital toxoplasmosis, necrosis of the brain tion may be difficult in hematoxylin and eosin–stained sections. The
is most intense in the cortex and basal ganglia and at times in the use of immunoperoxidase staining markedly improves the identifica-
periventricular areas.8,168 The necrotic areas may calcify and lead to tion of both tissue cyst and tachyzoite forms and highlights the pres-
striking radiographic findings suggestive but not pathognomonic of ence of T. gondii antigens (see Fig. 280-3B).29 T. gondii DNA can be
toxoplasmosis. Hydrocephalus may result from obstruction of the amplified from cerebrospinal fluid (CSF) or brain biopsy specimens of
aqueduct of Sylvius or foramen of Monro. Tachyzoites and tissue cysts patients with TE.173 Of note, PCR-positive results in brain biopsy speci-
may be seen in and adjacent to necrotic foci, near or in glial nodules, mens need to be interpreted with caution. These results may be positive
in perivascular regions, and in cerebral tissue uninvolved by inflam- in patients chronically infected with the parasite whose CNS pathology
matory change.169 The necrotic brain tissue autolyzes and is gradually can be explained by a diagnosis other than TE.
shed into the ventricles. The protein content of such ventricular fluid At autopsy in AIDS patients with TE, there is almost universal
may be in the range of grams per deciliter and has been shown to involvement of the cerebral hemispheres and a remarkable predilec-
contain significant amounts of T. gondii antigens. tion for the basal ganglia.86 In a consecutive autopsy study of 204
The presence of multiple brain abscesses is the most characteristic patients who died of AIDS, 46 (23%) had morphologic evidence of
feature of TE in severely immunodeficient patients and is particularly cerebral toxoplasmosis.172 In 38 (83%) of the 46 cases, histologic evi-
characteristic in patients with AIDS.6,170 Brain abscesses in AIDS dence of toxoplasmosis was restricted to the CNS. The cerebral hemi-
patients are characterized by three histologic zones. The central area is spheres were affected in 91% of cases and the rostral basal ganglia
avascular. Surrounding this is an intermediate hyperemic area with a in 78%.
prominent inflammatory infiltrate and perivascular cuffing by lympho- A “diffuse form” of TE has been described with histopathologic
cytes, plasma cells, and macrophages. Many tachyzoites and, at times, findings of widespread microglial nodules without abscess formation
tissue cysts as well, appear at the margins of necrotic areas. An outer in the gray matter of the cerebrum, cerebellum, and brainstem.174 In

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3130
these patients, involvement by T. gondii was confirmed by immuno- predominating.189 Focal necrosis with edema and an inflammatory
peroxidase stains that demonstrated tissue cysts and tachyzoites. In infiltrate is typical,188 although abscesses may also be noted.188,189
diffuse TE, the clinical course progresses rapidly to death. It has been Similar histologic findings are seen in the non-AIDS immunodeficient
postulated that in such cases, the lack of characteristic findings on population,6 and in both groups cardiac myocytes may be packed with
Part III  Infectious Diseases and Their Etiologic Agents

computed tomography (CT) or magnetic resonance imaging (MRI) tachyzoites (to produce pseudocysts) in the absence of an inflamma-
studies is due to insufficient time for abscesses to form before death tory response.
occurs. Biopsy-proven toxoplasmic myocarditis and polymyositis in the
Leptomeningitis is infrequent and, when present, occurs over adja- setting of acute toxoplasmosis have been reported in otherwise
cent areas of encephalitis. Spinal cord necrotizing lesions are seen at immunocompetent individuals and in patients on corticosteroids
autopsy in approximately 6% of patients with TE. The differential diag- (see Fig. 280-3C and D).164
nosis of TE lesions includes CNS lymphoma, progressive multifocal
leukoencephalopathy, and infection caused by organisms such as cyto- Other Organ Systems
megalovirus (CMV), Cryptococcus neoformans, Aspergillus spp., and Extensive involvement of the gastrointestinal tract in AIDS patients
M. tuberculosis. More than one agent may be present. may occur with tremendous variation in the inflammatory
response.190,191 Hemorrhagic gastritis and colitis have been described.192
Lung Other organs reported to be involved during toxoplasmosis include
Pulmonary toxoplasmosis in the immunodeficient patient may appear the liver,193 pancreas,194 seminiferous tubules,195 prostate,195 adrenals,196
in the form of interstitial pneumonitis, necrotizing pneumonitis, con- kidneys,197 and bone marrow.198
solidation, pleural effusion, empyema, or all of these.175 The pneumo-
nitis is associated with the development of fibrinous or fibrinopurulent CLINICAL MANIFESTATIONS
exudate. Tachyzoites may be found in alveolocytes, alveolar macro- Toxoplasmosis describes the clinical or pathologic disease caused by T.
phages, pleural fluid, or extracellularly within alveolar exudate. T. gondii and is distinct from T. gondii infection, which is asymptomatic
gondii DNA may be demonstrated in bronchoalveolar lavage (BAL) in the vast majority of immunocompetent patients.
fluid by the PCR.176 Toxoplasmosis is conveniently classified into five categories: (1)
acquired in the immunocompetent patient, (2) acquired or reactivated
Eye in the immunodeficient patient, (3) ocular, (4) in pregnancy, and (5)
Chorioretinitis in AIDS patients is characterized by segmental panoph- congenital. In any category, the clinical presentations are not specific
thalmitis and areas of coagulative necrosis associated with tissue cysts for toxoplasmosis, and a wide differential diagnosis must be considered
and tachyzoites.177 Numerous organisms in the absence of remarkable for each clinical syndrome. Furthermore, methods of diagnosis and
inflammation may be seen around thrombosed retinal vessels adjacent interpretation of test results may differ for each clinical category. Sero-
to necrotic areas. Multiple and bilateral lesions may occur.177 Amplifi- logic test results consistent with an infection acquired in the distant
cation of parasite DNA in both aqueous humor and vitreous fluid has past for a nonimmunocompromised pregnant woman in her first half
confirmed or supported the diagnosis of toxoplasmic chorioretinitis in of pregnancy are interpreted as no risk for congenital toxoplasmosis,
patients with atypical retinal findings for ocular toxoplasmosis or who whereas the same results for a patient about to undergo an allogeneic
are immunocompromised.178,179 hematopoietic stem cell or bone marrow transplant are interpreted as
Eye infection in immunocompetent patients produces acute cho- high risk for life-threatening toxoplasmosis in the post-transplant
rioretinitis characterized by severe inflammation and necrosis.177 period, particularly if the patient develops graft-versus-host disease
Granulomatous inflammation of the choroid is secondary to the (GVHD).
necrotizing retinitis. There may be exudation into the vitreous or
invasion of the vitreous by a budding mass of capillaries. Although Toxoplasmosis in the
rare, tachyzoites and tissue cysts may be demonstrated in the retina. Immunocompetent Patient
The pathogenesis of recurrent chorioretinitis is controversial. One In the United States and Europe, only 10% to 20% of cases of T. gondii
school proposes that rupture of tissue cysts releases viable organisms infection in immunocompetent adults and children are symptom-
that induce necrosis and inflammation, whereas another school atic.199 Recent reports suggest that this proportion may be higher in
contends that chorioretinitis results from a hypersensitivity reaction other areas of the world, such as Brazil and other countries in South
triggered by unknown causes.177 A study demonstrating efficacy of America.181,184 In addition, it appears that disease severity can also be
trimethoprim-sulfamethoxazole (TMP-SMX) in preventing recur- greater in countries outside Europe and the United States. For instance,
rences of chorioretinitis is consistent with the hypothesis that active community outbreaks of acute toxoplasmosis with an unusually severe
organism replication is necessary for recurrence.180 clinical presentation have been recently reported from Suriname and
Recent studies have revealed a much higher incidence of ocular French Guiana.200,201 In two reports, immunocompetent patients pre-
disease, which is often severe, among infected immunocompetent sented with severe disseminated disease, including pneumonia and
persons in South America than in North America or Europe.181-184 This hepatitis, that resulted in four deaths, including one newborn and one
difference seems most likely to be due to differences in the strains of fetus, among 22 patients. Based on genotype analysis with microsatel-
Toxoplasma that predominate in these different regions.17 This is con- lite markers, “atypical” strains (i.e., not one of the three major strains
sistent with observations within the United States, where specific seen in Europe and North America) were responsible for these out-
strains appeared to be associated with severe disease, although these breaks. The remarkable differences in clinical presentation of toxoplas-
studies involved relatively few patients and cannot be considered mosis among patients from various regions of the world have significant
definitive.185 implications when generating a differential diagnosis in travelers from
the United States and Europe who become ill.
Skeletal and Heart Muscle When clinical manifestations are present, toxoplasmosis most often
Myositis caused by T. gondii has been reported in as high as 4% of manifests as painless cervical lymphadenopathy, but any or all lymph
HIV-infected patients who present with neuromuscular symptoms, node groups may be enlarged. On palpation, the nodes are usually
and the same percentage has been observed in autopsy series of AIDS discrete and nontender, rarely more than 3 cm in diameter, may vary
patients in whom a systematic histologic evaluation of the skeletal in firmness, and do not suppurate.202 However, the nodes may be occa-
muscle was performed.186 Successful isolation from skeletal muscle sionally tender or matted. Fever, malaise, night sweats, myalgias, sore
biopsies has been reported.187 Microscopy has revealed necrotic muscle throat, arthralgias, maculopapular rash, hepatosplenomegaly, or small
fibers with a variable inflammatory reaction. Skeletal muscle involve- numbers of atypical lymphocytes (less than 10%) may be present.
ment has also been reported in the non-AIDS immunodeficient The clinical picture may resemble infectious mononucleosis or CMV
patient.6,164 infection, but toxoplasmosis probably causes no more than 1% of
Toxoplasmic myocarditis is frequently noted at autopsy in AIDS “mononucleosis” syndromes.203 Retroperitoneal or mesenteric lymph-
patients but is usually clinically inapparent,188 with CNS manifestations adenopathy may produce abdominal pain accompanied by diarrhea.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3131
Neurocognitive abnormalities appear to be common during the acute index of suspicion because routine laboratory tests to detect bacterial,
infection among immunocompetent patients, according to a recent viral, or fungal infections will not alert clinical laboratory personnel
report.204 or the clinician to the presence of T. gondii as the etiologic agent.
Toxoplasmic chorioretinitis as a manifestation of acute acquired T-cell–mediated immunity defects appear to confer the highest risk for

Chapter 280  Toxoplasma gondii


infection is more common than previously recognized.205-207 Chorio- toxoplasmosis as observed in patients with hematologic malignancies
retinitis in the setting of acute acquired toxoplasmosis can occur either (especially Hodgkin’s disease and other lymphomas), organ transplant
sporadically or in the context of an epidemic of acute toxoplasmo- recipients, or AIDS and those receiving immunosuppressive therapy
sis.206,208 For further discussion of this clinical entity, see “Ocular Toxo- with high doses of corticosteroids or immunomodulators, such as anti–
plasmosis in Immunocompetent Patients.” TNF-α agents, for instance, natalizumab or alemtuzumab.220 In immu-
In most cases, the clinical course of toxoplasmosis in the immuno- nodeficient patients, encephalitis, pneumonitis, and myocarditis reflect
competent patient is benign and self-limited. Symptoms, if present, active infection in the most commonly involved organs.6 Pneumonitis
usually resolve within a few months and rarely persist beyond 12 is a common and underrecognized manifestation of toxoplasmosis in
months. Lymphadenopathy may wax and wane for months and, in these patients. Fever of unknown origin may be the sole manifestation
unusual cases, for 1 year or longer. Rarely, an apparently healthy person of toxoplasmosis in the early stages of the disease. Disseminated infec-
develops clinically overt disease, for instance, fever of unknown origin tion with multiorgan involvement is not unusual; clinical manifesta-
or potentially fatal disseminated disease, with myocarditis, pneumoni- tions may not necessarily reflect the extent and severity of the
tis, hepatitis, or encephalitis. In children, nephrotic syndrome has been disseminated infection. Mortality approaches 100% if the infection is
reported in association with acute toxoplasmosis.209 These more aggres- not treated or is treated only late in its course. Whereas serious toxo-
sive forms of the disease have been more commonly reported from plasmosis in these patients often reflects recrudescence of a latent
South America.200,201 None of the clinical presentations of acquired infection acquired in the distant past (as observed in the setting of
toxoplasmosis is distinctive; the differential diagnosis of TL includes AIDS or bone marrow transplantation), it may also result from recently
lymphoma, infectious mononucleosis, CMV or human herpesvirus 6 acquired acute infection (as observed in solid-organ transplants
(HHV-6) “mononucleosis,” cat-scratch disease, sarcoidosis, tuberculo- through the transplanted organ) or, more rarely, through the oral route.
sis, tularemia, metastatic carcinoma, and leukemia. Acute acquired Although clinical manifestations are similar in patients with different
toxoplasmosis associated with multiorgan involvement has been causes for their immunosuppression, additional considerations are
reported to mimic other causes of pneumonitis, hepatitis, myocarditis, provided here for the organ transplant recipient and patient with AIDS.
polymyositis, or fever of unknown origin in apparently immunocom-
petent patients.199 Toxoplasmosis in the Solid-Organ
T. gondii has been estimated to cause 3% to 7% of clinically signifi- Transplant Patient
cant lymphadenopathy.202 The major diagnostic confusion with toxo- Patients with solid-organ transplants will develop toxoplasmosis most
plasmic lymphadenopathy occurs with Hodgkin’s disease and the commonly as a result of acquiring T. gondii infection through the
lymphomas. The diagnosis of recently acquired toxoplasmic lymph- transplanted organ, when the allograft of a seropositive donor (D+) is
adenopathy is easily made serologically, but unfortunately, physicians given to a seronegative recipient (R−), resulting in a D+/R− mismatch
often do not consider this diagnosis in patients with lymphadenopathy. (Table 280-1). Toxoplasmosis can also be the result of reactivation of
Serologic test titers diagnostic of acute T. gondii infection are often a previously acquired infection in the recipient, regardless of the sero-
obtained after histologic examination of a biopsied node has suggested logic status of the donor (D−/R+ or D+/R+, see Table 280-1). Fever is
the possibility of toxoplasmosis.210 often the first manifestation in transplant recipients, followed by signs
Myocarditis as a manifestation of acute toxoplasmosis has been referable to the brain and lungs.
reported in relatively few patients.164,211,212 It may occur clinically as an Knowledge of the overall prevalence of Toxoplasma antibodies in a
isolated disease process or as part of a variety of manifestations of dis- population does not accurately predict the percentage of D+/R− T.
seminated infection. Manifestations include arrhythmias, pericarditis, gondii mismatches. Rather, this will depend on the prevalence of T.
and heart failure.164 gondii antibodies in the age groups of the donor and recipient popula-
Myositis resembling polymyositis as a manifestation of acute toxo- tions. For example, in a given geographic area, the prevalence of anti-
plasmosis has also been reported infrequently.164,213,214 Dermatomyosi- bodies in young heart donors may be 3% to 10%, whereas in the older
tis has been associated with toxoplasmosis, although a cause-and-effect population of individuals who would more likely be recipients, it may
relationship has not been proved.215,216 be 15% to 30%. Testing for Toxoplasma IgG antibodies should be per-
The clinical features of toxoplasmic myocarditis and polymyositis formed in every organ transplant candidate before transplantation and
are illustrated by a case in which both were present in the same indi- on serum from every organ donor. This will allow identification of
vidual.164 A 43-year-old woman presented with cardiogenic pulmonary those recipients at greatest risk of developing toxoplasmosis either
edema, followed by progressive sinus bradycardia and subsequent because they were seronegative before transplantation and received an
complete heart block; viral myocarditis was considered the most likely organ from a seropositive individual or because they were seropositive
diagnosis. During the ensuing months, she developed proximal muscle
weakness while being treated with corticosteroids; an endomyocardial
biopsy (see Fig. 280-3C) and a quadriceps muscle biopsy (see Fig. 280-
TABLE 280-1  Source of Toxoplasmosis in the
3D) revealed T. gondii.164 Her symptoms improved on pyrimethamine-
Organ Transplant Patient
sulfadiazine. One year after her initial presentation with myocarditis,
retinal lesions characteristic of toxoplasmic chorioretinitis were Transplant of an Infected Organ to a Seronegative Recipient
observed in her right eye. Serologic test results and follow-up were (D+R−)
consistent with recently acquired toxoplasmosis.164 Heart
Several epidemiologic studies have suggested an association Heart-lung
between infection with T. gondii and schizophrenia and other mental Kidney
illnesses but a definitive etiologic role of the parasite in such disorders Liver and liver/pancreas
has not been established.217-219 Population based studies that include Bone marrow (rare)
large cohorts of patients followed since gestation will be necessary to Reactivation of Latent Infection in a Seropositive Recipient
clarify this potential association. (D−R+ and D+R+)
Bone marrow
Toxoplasmosis in the  
Hematopoietic stem cell
Immunodeficient Patient
Liver
In immunocompromised patients toxoplasmosis can present with a
wide spectrum of clinical manifestations. Disseminated disease has a Kidney (rare)
100% case-fatality rate if untreated. Early diagnosis requires a high D, donor; R, recipient.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3132
before transplantation and thus are at risk for reactivation of their renal allografts from the same donor has occurred. Chorioretinitis has
latent (chronic) Toxoplasma infection. Unfortunately, Toxoplasma been reported as the presenting manifestation of toxoplasmosis in a
serologies obtained in the early months post-transplantation frequently kidney transplant patient.227
are not helpful, even in the presence of serious toxoplasmosis in these
Part III  Infectious Diseases and Their Etiologic Agents

patients. T. gondii antibodies demonstrable before transplantation Liver Transplantation


might become negative, rise, or show no change post-transplantation After orthotopic liver transplantation, toxoplasmosis most often results
despite life-threatening toxoplasmosis.221 Transfusion may further from activation of a latent infection in the transplanted allograft,228 but
compound the difficulties encountered in serodiagnosis. it occurs as well from activation of a quiescent pretransplantation infec-
The incidence of toxoplasmosis among various organ transplant tion in the recipient. In most published cases, clinical manifestations of
recipients managed at 11 tertiary care hospitals in Spain between 2000 toxoplasmosis appeared within the first 3 months post-transplantation.
and 2009 was 0.14% (22/15,800).222 The incidence was significantly Fever was usually the first manifestation, and pneumonia, meningitis/
greater in heart compared with kidney and liver recipients. The only encephalitis, and multiorgan failure were frequently observed. Retino-
independent risk factor identified in multivariate analysis was negative choroiditis requiring enucleation was observed in one patient.229
serostatus before transplant. Overall mortality was 14%, although two Although it is a rare event, toxoplasmosis in this population is most
of the three deaths occurred in untreated patients who were diagnosed often fatal.230
at autopsy.
At autopsy, histopathologic evidence of multiorgan involvement Toxoplasmosis in the Bone Marrow
has been observed, most commonly of the brain, heart, and lungs but Transplant and Hematopoietic Stem
also including the eyes, liver, pancreas, adrenal, and kidney. Cell Transplant Recipient
Donor bone marrow from the patient (autologous) or a matched
Heart Transplantation related or unrelated donor (allogeneic) used to be the only source of
In a review of infections in cardiac transplant recipients at Stanford cells for bone marrow transplant (BMT) recipients. Stem cell trans-
Medical Center from 1980 to 1996, results of serologic testing for plantation is now also done using stem cells harvested from the periph-
Toxoplasma were available for 582 donors (35 [6%] had T. gondii– eral blood (hematopoietic stem cell transplant [HSCT]) of the patient
specific IgG antibodies) and 607 recipients (98 [16%] were positive).223 (autologous), matched related or unrelated donor (allogeneic), matched
Results of serologic testing for Toxoplasma were available for 575 D/R parent (haploid), or less commonly, umbilical cord blood. To keep this
pairs; of these, 454 (79%) were D−/R−, 84 (14.6%) D−/R+, 32 (5.6%) D+/ literature separate, HSCT and BMT are presented separately in this
R−, and 5 (0.8%) D+/R+. Of the 32 D+/R− patients, 16 were receiving section. A review of 41 cases of toxoplasmosis in patients who had
TMP-SMX and/or pyrimethamine prophylaxis, and none developed undergone HSCT in 15 European transplantation centers, from 1994
toxoplasmosis; however, 4 (25%) of the 16 D+/R− patients who were not through 1998, found no cases among 6787 autologous HSCT, whereas
taking either TMP-SMX or pyrimethamine developed toxoplasmosis, it occurred in 0.97% of 4231 allogeneic transplants.231 The relatively
and all died of the infection. None of the 98 patients who were sero- low number of cases in their large survey is likely due to the use of
positive for T. gondii preoperatively developed clinical evidence of TMP-SMX prophylaxis after engraftment in all allogeneic BMTs in
reactivation of the infection. The importance of prophylaxis is further 91% of the institutions. Of the patients with available serologies before
evidenced from an earlier study at Papworth Hospital in England. Fatal transplantation, 94% were seropositive for T. gondii. GVHD had devel-
or severe toxoplasmosis developed in 57% (4/7) of D+R− mismatched oped before toxoplasmosis in 73%. Thirty patients (73%) had not
heart transplant patients not receiving prophylaxis. Use of pyrimeth- received prophylaxis for toxoplasmosis, and only 3 were receiving it at
amine, 25 mg/day for 6 weeks reduced the transmission rate to 14% the time of disease onset. Median time of onset was day 64 (range, 4
(5/37). In those patients who received pyrimethamine and were to 516 days). Fever with neurologic or pulmonary symptoms was seen
infected by the donor heart, only 1 (20%) developed symptoms of the most commonly; myocarditis was frequently seen at autopsy. Six
infection in contrast to 4 of 4 (100%) who did not receive pyrimeth- patients had fever without evidence of organ involvement. Twenty-two
amine prophylaxis. Subsequently, prophylaxis with TMP-SMX (480 mg (63%) died of toxoplasmosis. It is noteworthy that of the 23 patients
twice daily orally for 1 year post-transplantation and when on oral who received specific therapy for toxoplasmosis for greater than or
prednisolone) was used in heart and lung transplant patients. Of those equal to 6 days, 11 (48%) had a complete response, and 3 (13%) others
who were alive at 3 months post-transplantation, 28 (8.75%) were T. improved. In 106 T. gondii–seropositive adult recipients of HSCTs, the
gondii mismatches; none had evidence of having acquired Toxoplasma incidence of reactivation of toxoplasmosis in the first 6 months after
infection. These investigators observed that use of prophylaxis might transplantation was prospectively studied.232 Toxoplasma serologies
prolong the period before observation of seroconversion of donor- had been obtained pretransplantation. The incidence of reactivation
acquired infection in heart transplant patients for as long as 14 months within 6 months after transplantation, as established by a positive PCR
post-transplantation.224,225 test result in peripheral blood, was observed in 16 of the 106 patients
Toxoplasmosis in heart transplant recipients may simulate organ (16%). Of the 16 patients with positive PCR test results, 6 (38%) devel-
rejection. In such cases, toxoplasmosis has frequently been diagnosed oped disease (toxoplasmosis). Thus, PCR may be useful in monitoring
by endomyocardial biopsy. at-risk patients (i.e., seropositive patients identified in the pretrans-
It is important to recognize that many heart transplant recipients plantation period) (see “Diagnosis”).
with T. gondii antibodies before transplantation may show increases Survival in this setting is highest in patients with ocular and/or
in T. gondii–specific antibodies (IgG and IgM). These patients have isolated cerebral toxoplasmosis, primarily when treatment is begun as
not necessarily developed a clinical illness that can be attributed to soon as the diagnosis is suspected.82,231 Survival in the presence of dis-
toxoplasmosis. seminated toxoplasmosis is rare in these HSCT patients.82,231 Although
reactivation of latent Toxoplasma infection in allogeneic HSCT recipi-
Kidney Transplantation ents most often occurs in the first 6 months post-transplantation (the
In a review of 31 cases of toxoplasmosis in renal transplant patients, majority occur in the first 30 to 90 days), late reactivation has been
the majority occurred within the first 3 months after transplantation; observed and must be considered in patients in whom late-onset
3 cases occurred more than 1 year after transplantation, and 9 occurred (beyond 6 months post-transplantation) GVHD occurs.232,233
during or immediately after a rejection episode.226 The greatest risk was The incidence of toxoplasmosis in BMT has been reported to range
in D+/R− mismatches. Fever, CNS signs, and pneumonia were the main from 0.3% to 5% and is influenced by the prevalence of pretransplanta-
clinical features. Chest radiographs showed bilateral pneumonia in tion antibodies and whether toxoplasmosis prophylaxis was used.
most cases. The most common organs involved in the 15 cases diag- Major risk factors for toxoplasmosis included the presence of pretrans-
nosed at autopsy were brain, heart, and lungs. T. gondii was not demon- plantion T. gondii antibodies in recipients and the occurrence of
strable in the kidneys. Whereas the overall mortality rate was 64%, 10 GVHD. In a review of 110 published cases of toxoplasmosis after
of 11 treated patients survived, emphasizing the importance of early BMT,82 96% occurred after allogeneic BMT. Onset post-transplantation
diagnosis and treatment. Acute toxoplasmosis in two recipients of occurred on days 1 to 30 in 13%, on days 31 to 100 in 64%, and on

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3133
days greater than 100 in 23%. The infection occurred primarily in may be the first sign; if pulmonary infiltrates are observed, especially
recipients who were seropositive pretransplantation (88%), meaning in the setting of rapid deterioration, immediate attempts at diagnosis
infection was more often reactivation in the recipient—not trans- must be made and empirical treatment begun. Undoubtedly, the high
planted from the donor. The diagnosis was made antemortem in only mortality in many instances has been due to lack of early diagnosis and

Chapter 280  Toxoplasma gondii


47% of the cases. Overall mortality was 80% (median, 87 days post- treatment. The organism can be observed on microscopic examination
transplantation), and in 66%, it was attributed to toxoplasmosis of BAL material. PCR on material obtained at BAL may be the diag-
(median, 74 days post-transplantation). Patients with isolated cerebral nostic procedure of choice.235 PCR also can be performed on blood,
involvement had a better outcome (58% survival) than patients with serum, CSF, and bone marrow aspirates.
disseminated toxoplasmosis (20% survival); underlying disease was the Ocular toxoplasmosis has been reported after allogeneic and autol-
only factor associated with clinical presentation, with acute leukemia ogous BMT and HSCT. In some cases, the ocular disease was due to
being more common in patients with disseminated disease. reactivation of a previously observed toxoplasmic chorioretinitis,
TMP-SMX prophylaxis, primarily used by transplantation teams to whereas in most, it has been associated with disseminated infection.
prevent Pneumocystis pneumonia, has been successful in prevention of Definitive diagnosis has been by direct observation of the parasite in
toxoplasmosis. In HSCT patients, this is usually begun after engraft- histopathology sections, culture of tissue samples, or by PCR on vitre-
ment because of the potential of the drug combination for bone marrow ous fluid.
suppression. The delay in instituting prophylaxis likely results in many
more cases of toxoplasmosis than would be expected to occur if ade- Toxoplasmosis in the AIDS Patient
quate prophylaxis was begun early after transplantation. This problem Clinical manifestations of toxoplasmosis in AIDS patients commonly
highlights the importance of PCR monitoring of at-risk patients and reflect encephalitis (i.e., TE), or infection of the lung (pneumonitis),
identifying additional drugs for prophylaxis in these patients.234 and the eye (chorioretinitis).236 Toxoplasmosis with multiorgan
Toxoplasmosis in BMT patients frequently involves the lung involvement manifesting with acute respiratory failure and hemody-
(usually in the setting of multiorgan disease), with a high associated namic abnormalities similar to septic shock has been reported,
mortality (>90%) (Fig. 280-4). Most patients die within 7 days of onset although septic shock has not been definitely proved to be due to
of pulmonary symptoms and often suffer acute respiratory distress T. gondii.237 TE is the most common presentation of toxoplasmosis in
syndrome. In a review of 25 cases of pulmonary toxoplasmosis in BMT, AIDS patients and is a frequent cause of focal CNS lesions in these
onset of symptoms referable to the lungs occurred from 7 days to patients.86 A wide range of clinical findings, including altered mental
1 year post-BMT; the majority occurred in the first 6 weeks.235 Fever state, seizures, weakness, cranial nerve disturbances, sensory abnor-

A 4/23/2012 4/24/2012 5/8/2012

B 4/24/2012 5/3/2012

FIGURE 280-4  Pulmonary toxoplasmosis. Serial chest radiographs (A) and computed tomography scans of the chest (B) of a patient who had
received a haplo-cord transplant as treatment for myelodysplastic syndrome complicating aplastic anemia and subsequently presented with pulmonary
symptoms. Pulmonary toxoplasmosis was diagnosed by polymerase chain reaction (PCR) of a bronchoalveolar lavage sample obtained on April 24, 2012.
The patient also had a positive PCR for toxoplasmosis in blood and cerebrospinal fluid. The patient responded to treatment with intravenous trimethoprim-
sulfamethoxazole and subsequently oral pyrimethamine-sulfadiazine-leucovorin, with resolution of pneumonia and conversion of PCR to negative.
(Courtesy Dr. Richard Childs, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD.)

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3134
malities, cerebellar signs, meningismus, movement disorders, and
neuropsychiatric manifestations are seen in TE. The characteristic pre-
sentation usually has a subacute onset with focal neurologic abnor-
malities in 58% to 89% of patients. However, in 15% to 25% of cases,
Part III  Infectious Diseases and Their Etiologic Agents

the clinical presentation may be more abrupt, with seizures or cerebral


hemorrhage. Most commonly, hemiparesis or abnormalities of speech,
or both, are the major initial manifestations. Brainstem involvement
often produces cranial nerve lesions, and many patients exhibit cere-
bral dysfunction with disorientation, altered mental state, lethargy, and
coma. Less commonly, parkinsonism, focal dystonia, rubral tremor,
hemichorea-hemiballismus, panhypopituitarism, diabetes insipidus, or
the syndrome of inappropriate antidiuretic hormone secretion may
dominate the clinical picture. In some patients, neuropsychiatric
symptoms, such as paranoid psychosis, dementia, anxiety, and agita-
tion, may be the major manifestations.
Diffuse TE174 has been reported in relatively few AIDS patients; its
actual incidence is unknown. This form of TE may manifest acutely A
and can be rapidly fatal; generalized cerebral dysfunction without focal
signs is the most common manifestation, and CT scans may be within
normal limits or reveal cerebral atrophy.
Spinal cord involvement by T. gondii in AIDS patients manifests as
motor or sensory disturbances of single or multiple limbs, bladder or
bowel dysfunctions (or both), and local pain. Patients may present with
a clinical syndrome resembling a spinal cord tumor. Reports of cervical
myelopathy,238 thoracic myelopathy,239 and conus medullaris syn-
drome240 have been published.
Pulmonary disease caused by toxoplasmosis has been reported in
patients with AIDS, and the diagnosis may be made by demonstration
of the parasite in BAL fluid.241 In France, before ART and routine
use of prophylaxis, the prevalence of pulmonary toxoplasmosis in
patients dually infected with HIV and T. gondii was estimated to be
approximately 5%.242 Pulmonary toxoplasmosis occurs mainly in
patients with advanced AIDS (mean CD4 count, 40 cells/mm3 ± 75 SD)
and primarily presents as a prolonged febrile illness with cough and B
dyspnea,241 which may be clinically indistinguishable from Pneumo­
cystis pneumonia. Mortality, even when treated appropriately, may
be as high as 35%. Extrapulmonary disease may be present in about
50% of cases with toxoplasmic pneumonitis.237 Often, pulmonary toxo-
plasmosis is not associated with TE; however, TE may develop after
successful treatment of pulmonary toxoplasmosis when therapy is dis-
continued. The differential diagnosis of toxoplasmic pneumonitis
includes Pneumocystis pneumonia, viral pneumonia (e.g., caused by
CMV or community-acquired respiratory viruses), pneumonia caused
by atypical pathogens (e.g., Chlamydia spp., Mycoplasma pneumoniae),
and infection with M. tuberculosis, Cryptococcus neoformans, Coccidi-
oides spp., and Histoplasma capsulatum.
Toxoplasmic chorioretinitis is seen relatively infrequently in AIDS
patients243; it commonly manifests with ocular pain and loss of visual
acuity (Fig. 280-5). Funduscopic examination usually demonstrates
necrotizing lesions that may be multifocal or bilateral.243 Overlying
vitreal inflammation is often present and may be extensive. The optic
nerve may be involved in as many as 10% of cases. Toxoplasmic cho- C
rioretinitis in AIDS patients has been associated with concurrent TE
FIGURE 280-5  Toxoplasmic chorioretinitis. A, Active chorioretinitis
in up to 63% of patients. The differential diagnosis of toxoplasmic with two lesions and vitreous haze, in a human immunodeficiency virus–
chorioretinitis in AIDS patients includes CMV retinitis, syphilis, infected patient. B, A large inactive macular lesion typical of congenital
herpes simplex, varicella zoster, and candidiasis. The diagnosis relies disease. C, Active chorioretinitis in an immunocompetent patient from an
primarily on clinical findings and the response to anti–T. gondii endemic region in Brazil. There is an inactive lesion on the right. Note the
therapy, although definitive diagnosis may be made by demonstration macular star, which is due to an exudate around the macula. (Courtesy Dr.
of the organism in retinal biopsies,243 isolation of the parasite from Robert Nussenblatt, National Eye Institute, National Institutes of Health,
vitreous aspirates,244 or amplification of the parasite DNA by PCR.178 Bethesda, MD; Dr. Claudio Silveira, Erechim, Brazil; and Dr. Rubens Belfort,
Gastrointestinal involvement may result in abdominal pain, ascites São Paulo Brazil.)
(resulting from involvement of the stomach, peritoneum, or pancreas),
or diarrhea. Acute hepatic failure caused by T. gondii has been accounting for 26% of such cases in one recent study, with a 1-year
reported,193 as has musculoskeletal involvement.186 estimated survival of 77%.246 In contrast to mycobacterial and crypto-
Although the incidence of toxoplasmosis in HIV-infected patients coccal infections, immune reconstitution inflammatory syndrome
began to decrease after the broad use of TMP-SMX for PCP prophy- after the initiation of ART has been only rarely reported in association
laxis, the incidence decreased dramatically as a result of the immune with toxoplasmosis.247-250 In patients with CD4 counts that are sus-
reconstitution associated with ART regimens. Approximately fourfold tained to levels greater than 200 cells/µL for 3 to 6 months while
decreases in both incidence and death have been reported after the receiving ART, prophylaxis regimens can be safely discontinued,
wide availability of ART regimens.87,88,245 Yet despite this, TE remains although rare cases of disease can occur even in patients with well-
one of the most common HIV-related neurologic disorders, controlled HIV infection.251-254

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3135
There are no data on whether the strain of T. gondii affects the that the congenitally acquired disease has a more guarded prognosis.
clinical outcome of infection in AIDS patients, and there are conflict- From the public health perspective, it is important epidemiologically
ing reports on whether a particular strain is more likely to cause infec- to establish whether the patient has acute acquired infection, to initiate
tion in such patients.13,255 efforts to identify the possible source of T. gondii infection and to

Chapter 280  Toxoplasma gondii


determine whether other individuals who may be at high risk for
Ocular Toxoplasmosis in developing severe, life-threatening disease (i.e., fetuses of serologically
Immunocompetent Patients negative pregnant women or immunodeficient individuals) shared the
T. gondii is one of the most frequently identified etiologies of uveitis same exposure as the individual with acute acquired toxoplasmic cho-
and the most commonly identified pathogen to infect the retina of rioretinitis. Serologic tests have been useful in establishing whether
otherwise immunocompetent individuals.256 It accounted for 8.4% of such patients have been infected recently.205,206 In patients with cho­
cases of uveitis seen in a clinic in San Francisco over a 24-year period.257 rioretinitis and IgG antibodies, additional serologic tests should be
Toxoplasmosis is responsible for more than 85% of posterior uveitis performed to determine whether the patient’s infection is recently
cases in southern Brazil, where, in one study, 9.5% of 21 seroconverters acquired.206
and 8.3% of 131 seropositive patients without ocular involvement T. gondii chorioretinitis may resemble the posterior uveitis of
developed typical lesions during 7 years of follow-up.258-260 Chorioreti- tuberculosis, syphilis, leprosy, or presumed ocular histoplasmosis
nal lesions may result from congenital or postnatally acquired infec- syndrome.
tion. In both these situations, lesions may occur during the acute or Atypical clinical and serologic manifestations of toxoplasmic cho-
latent (chronic) stage of the infection.206,261 Recurrences are frequent, rioretinitis have been reported most commonly in elderly and in
occurring in 79% of patients followed for more than 5 years in one immunodeficient individuals.178,266 Patients are considered to have
study, with a median time to recurrence of 2 years; they tend to occur atypical-appearing lesions when one or more of the following features
in clusters, that is, immediately after an episode rather than randomly, are present: multiple foci of active retinitis, acute retinal necrosis
are more common in patients older than 40 years, are more common syndrome (vitritis, peripheral retinitis, retinal vasculitis), significant
in the eye originally involved, and may be more common after cataract intraretinal hemorrhage, an absence of ophthalmoscopically visible
extraction.262-264 chorioretinal scarring. In patients with atypical lesions or an inade-
It is frequently difficult to determine whether the original infection quate clinical response to anti-Toxoplasma therapy or in whom other
was congenital or acquired in patients who suffer recurrences of cho- diagnostic procedures have not proved helpful, obtaining vitreous or
rioretinitis.207 Although previously it was thought that less than 1% of aqueous fluid (in some cases indicated for therapeutic reasons as well)
cases were acute infections, the remainder being reactivations, recent for PCR should be considered early in the workup (see “Diagnosis”).179
evidence indicate that 11.7% of U.S. ocular infections are acute.264a In the future, it may be possible to routinely determine which strain
Patients who present with chorioretinitis as a late sequela of the infec- of the parasite is responsible for the infection and make a more accu-
tion acquired in utero tend to have more severe disease and are more rate prognosis. Preliminary indications are that strain type may play
frequently in the second and third decades of life (it is rare after the an important part in determining the course of the infection,184,185 and
age of 40 years); bilateral disease, old retinal scars, and involvement of methods to distinguish strain type by looking for strain-specific anti-
the macula are hallmarks of the retinal disease in these cases, as are bodies using polymorphic peptides have shown promise.75,267
recurrences (see Fig. 280-5).8 By contrast, patients who present with
toxoplasmic chorioretinitis in the setting of acute toxoplasmosis are Toxoplasmosis during Pregnancy
more often between the fourth and sixth decades of life, most often As in other immunocompetent individuals, acute Toxoplasma infec-
have unilateral involvement, and have eye lesions that usually spare the tion is asymptomatic in the majority of pregnant women. Based on
macula and do not present with associated old scars.206,207 newly developed serologic assays that can detect antibodies to sporo-
Whereas acquired T. gondii infection in otherwise healthy adults is zoites,49 the majority of women in one U.S. study appeared to have been
most often subclinical, toxoplasmic chorioretinitis in these individuals initially infected with oocysts, and a high proportion were unable to
may result in complete or partial loss of vision or in glaucoma, and identify the potential source of such infection.268 Given these factors,
it may necessitate enucleation.8,265 Acute chorioretinitis may produce women should be universally screened by serology to maximize
symptoms of blurred vision, scotoma, pain, photophobia, and epiph- diagnosis of toxoplasmosis during pregnancy. The most commonly
ora. Impairment or a loss of central vision occurs when the macula is recognized clinical manifestation of recent infection is regional lymph-
involved. As inflammation resolves, vision improves, frequently adenopathy. The primary concern is transmission of infection to the
without complete recovery of visual acuity, caused in large part by fetus. The risk to the fetus does not correlate with whether the infection
macular scar formation.257 In most cases, toxoplasmic chorioretinitis in the mother was symptomatic or asymptomatic during gestation.
is diagnosed by ophthalmologic examination, and empirical therapy Transmission to the fetus has been limited almost solely to those
directed against the organism is often instituted based on clinical find- women who acquire the infection during gestation. Otherwise healthy
ings and serologic test results. Typical features of toxoplasmic chorio- women with prior Toxoplasma infection are protected from transmit-
retinitis include intensely white focal lesions with an overlying, intense, ting the infection to their fetuses. Rare exceptions to this dictum have
vitreous inflammatory reaction (see Fig. 280-5). Focal necrotizing been observed. In immunocompetent women infected with T. gondii
retinitis initially appears in the fundus as a yellowish-white, elevated shortly before conception, transmission to the fetus has occurred; in
cotton patch with indistinct margins, usually on the posterior pole. The these very rare instances, the acute infection was acquired within 3
lesions are often in small clusters, and individual lesions in the cluster months of conception.269-271 Transmission to the fetus has been rarely
may be of varied ages. With healing, the lesions pale, atrophy, and recognized as a consequence of reactivation of latent T. gondii infection
develop black pigment (see Fig. 280-5). There can also be an associated, in immunocompromised women infected with T. gondii before con-
secondary iridocyclitis and increased intraocular pressure.265 The ception (chronic infection) (e.g., pregnant women coinfected with HIV
classic “headlight in the fog” appearance is due to the presence of active and T. gondii,272 patients with systemic lupus erythematosus who are
retinal lesions with severe vitreous inflammatory reaction. The choroid being treated with corticosteroids). In addition, reinfection with a
is secondarily inflamed. Recurrent lesions tend to occur at the borders second, more aggressive strain of T. gondii of otherwise healthy preg-
of chorioretinal scars, and scars are often found in clusters. Panuveitis nant women who are already chronically infected with the parasite has
may accompany chorioretinitis, but isolated anterior uveitis has never been proposed and documented as a possible mechanism of transmis-
been proved to occur. sion to the fetus.109,269
Although the morphology of the lesions of acute toxoplasmic cho-
rioretinitis in the setting of postnatally acquired disease may be indis- Congenital Toxoplasmosis
tinguishable from those observed in patients who suffer acute eye Congenital infection may present in one of five forms: (1) ultrasound
disease in later life caused by a congenitally acquired infection, it is abnormalities consistent with toxoplasmosis or positive amniotic fluid
important to attempt to establish which type of the infection (postna- PCR test results in the fetus; (2) neonatal disease; (3) disease (mild or
tally acquired or congenital) is occurring in a given patient.206 It appears severe) occurring in the first months of life; (4) sequelae or relapse of

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3136
a previously undiagnosed infection during infancy, childhood, or ado- caused by organisms such as herpes simplex virus, CMV, and rubella
lescence; and (5) subclinical infection. virus. Signs include chorioretinitis, strabismus, blindness, epilepsy,
Data accumulated from prospective studies indicate that the inci- psychomotor or mental retardation, anemia, jaundice, rash, petechiae
dence and severity of congenital toxoplasmosis vary with the trimester resulting from thrombocytopenia, encephalitis, pneumonitis, micro-
Part III  Infectious Diseases and Their Etiologic Agents

during which the infection was acquired by the mother.8 Moreover, cephaly, intracranial calcification, hydrocephalus, diarrhea, hypother-
there is an inverse relationship between the frequency of transmission mia, and nonspecific illness.8 There may be no sequelae, or sequelae
and the severity of disease. Infants born of mothers who acquire their may develop or be evident at various times after birth.
infection in the first and second trimester more frequently show severe A detailed examination by an experienced clinician may be neces-
congenital toxoplasmosis.273 In contrast, the majority of children born sary to detect signs of the infection.8 In one prospective study,287 210
of women who acquire their infection during the third trimester are congenitally infected infants were identified: 2 patients (0.9%) died, 21
born with the subclinical form of the infection. However, if left (10.9%) had severe disease, 71 (33.8%) were mildly afflicted, and 116
untreated, as many as 85% of these latter children develop signs and (54.4%) were without signs of the infection. More intensive examina-
symptoms of the disease, in most cases chorioretinitis or delays in tion of the latter 116 infants revealed abnormalities in 39; abnormal
development.274,275 Although the majority of neonates born to mothers CSF was detected in 22 infants, chorioretinitis was seen in 17, and
infected later in gestation have subclinical infection, recent reports intracranial calcifications were found in 10. Premature infants often
suggest that when infected with a more virulent or atypical strain, suffer CNS disease and ocular disease in the first 3 months of life. Full-
severe congenital disease may occur despite having acquired their term infants frequently develop a milder disease manifested by hepa-
infection late in gestation.276 tosplenomegaly and lymphadenopathy that usually appear in the first
Infection acquired in the first trimester by women who were not 2 months of life. In these infants, disease reflecting damage to the CNS
treated with anti–T. gondii drugs resulted in congenital infection in 25% may occur later, and eye disease may occur months to years after birth.
of cases in one report.273 For second- and third-trimester infections, the Most untreated infants with subclinical infection at birth subse-
incidences of fetal infection were 54% and 65%, respectively.273 The quently develop signs or symptoms of congenital toxoplasmosis,
overall rate of vertical transmission in this study of untreated women including chorioretinitis, which may lead to blindness, neurologic
was 50%.273 Early treatment of the mother with spiramycin appears to manifestations such as seizures, and sensorineural hearing loss.275,288
reduce the incidence of congenital infection by about 60%.273,277,278-280 Prospective observational studies suggest that early initiation of spe-
Maternal infection acquired around the time of conception and within cific therapy (prenatally and postnatally) in infants with congenital
the first 2 weeks of gestation and treated with spiramycin usually does infection but without clinical signs will markedly reduce untoward
not result in transmission.279 Because of the high transmission rates sequelae.289,290 In one cohort, among children who were diagnosed with
observed in the late second trimester and during the third trimester, it toxoplasmic chorioretinitis only after their first year of life (thus, who
is recommended that pyrimethamine-sulfadiazine be used in patients were not treated during their first year of life), new chorioretinal
in whom acute infection is highly suspected or confirmed as having lesions were detected in more than 70%,291 whereas among 108 con-
occurred at 18 weeks of gestation or later. genitally infected children treated during their first year of life, only
Frequency of transmission to the fetus and severity of disease in the 31% developed at least one new chorioretinal lesion. Thirteen percent
offspring appear to be significantly affected by drug treatment with had occurrences when they were 10 years or older, indicating that
spiramycin and pyrimethamine-sulfadiazine as well. In cohorts where long-term follow-up is important.289,292 It appears that, when treated,
most of the mothers have been treated during gestation, transmission initially asymptomatic congenital toxoplasmosis has an overall good
rates were low in the first trimester. (i.e., 6% [95% confidence interval prognosis.289
{CI}, 3% to 9%)] at 13 weeks) and increased as expected with advanc- Uncommonly, latent T. gondii infection may reactivate in HIV-
ing gestational age (i.e., 40% [95% CI, 33% to 47%] at 26 weeks and infected women and result in congenital transmission of the parasite.
72% [95% CI, 60% to 81%] at 36 weeks)281 The overall rate of vertical Congenital toxoplasmosis appears to occur more frequently in the
transmission in this study of treated women was 29% (95% CI, 25% offspring of women infected with both HIV and T. gondii than in those
to 33%).281 Clinical signs in the infected infant were more likely of women who are infected with T. gondii but not with HIV.293 Infants
observed in offspring of women whose infection was acquired early in with congenital toxoplasmosis born to HIV-infected mothers are also
gestation. Depending on when during gestation the mother acquired infected with HIV, suggesting that factors predisposing to the vertical
her infection, the risk for severity of clinical manifestations in an transmission of HIV also favor the transmission of T. gondii, or vice
infected fetus was 61% (95% CI, 34% to 85%) at 13 weeks, 25% (95% versa. Congenital toxoplasmosis in the HIV-infected infant appears to
CI, 18% to 33%) at 26 weeks, and 9% (95% CI, 4% to 17%) at 36 run a more rapid course than that in the non–HIV-infected infant, with
weeks.281 A recent study reported the lowest rates of overall trans­ the development of failure to thrive, fever, hepatosplenomegaly, cho-
mission (4.8%) and clinical manifestations in the newborn (1.6%) rioretinitis, and seizures. Most children have multiorgan involvement,
published to date.282 In this cohort, pregnant women with T. gondii including CNS, cardiac, and pulmonary disease.
infection acquired during gestation received spiramycin until the 16th It has been recently reported that T. gondii causes more severe
week of gestation, followed by at least 4 weeks of a pyrimethamine- ocular disease in congenitally infected children in Brazil when com-
sulfadiazine–folinic acid combination independent of the infection pared with those in Europe.182 As stated earlier, this may be explained
status of the fetus. If fetal infection was suspected (e.g., ultrasound by the fact that pregnant women in Brazil are not routinely screened
abnormalities suggestive of congenital infection are observed) or con- and treated for toxoplasmosis during gestation and possibly that more
firmed (e.g., positive amniotic fluid PCR test), combination treatment virulent genotypes of the parasite predominate in Brazil but are rarely
was continued until delivery. In their cohort, their rates of transmission found in Europe.
for the first, second, and third trimester were 1.3%, 10.6%, and 21.7%, Congenital toxoplasmosis must be differentiated from rubella virus,
respectively. Another study reported that prenatal treatment with spi- CMV, herpes simplex virus, HHV-6, parvovirus B19, and lymphocytic
ramycin and/or pyrimethamine-sulfadiazine resulted in a significantly choriomeningitis virus infections; syphilis, listeriosis, and other bacte-
reduced risk of severe neurologic sequelae or death in the infected rial infections; other infectious encephalopathies; erythroblastosis
offspring.283 Of note, recently severe congenital toxoplasmosis has only fetalis; and sepsis. Herpes simplex virus, CMV, rubella virus, and syph-
been reported from countries such as the United States and Brazil, ilis may cause chorioretinitis; both CMV and rubella have been associ-
where universal screening and treatment during gestation have not ated with hydrocephalus, microcephaly, and cerebral calcification. A
been implemented.284,285 markedly elevated CSF protein concentration is a hallmark of congeni-
In addition to gestational age and treatment, transmission rates and tal toxoplasmosis.
severity of congenital disease are also likely to be affected by the strain T. gondii infection acquired during pregnancy has been implicated
of the parasite, infecting form of the parasite (cyst vs. oocyst), parasite in spontaneous abortion, stillbirth, and premature births. On rare
load,286 and immune status and genetics of the host. occasions, T. gondii has been isolated from the abortuses of women
Clinical manifestations of congenital toxoplasmosis vary. Most with chronic infection, but the frequency of T. gondii infection as a
signs and clinical presentations are nonspecific and may mimic disease cause of abortion is unknown and controversial.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3137
As with ocular disease, the strain of T. gondii may have an impact successfully by the Palo Alto Medical Foundation–Toxoplasma Serol-
on the outcome of congenital infection. Studies in Europe have revealed ogy Laboratory (PAMF-TSL, 650-853-4828, www.pamf.org/serology/)
conflicting data on whether type I strains, in particular, might be more to determine whether serologic test results are more likely consistent
likely to be responsible for congenital infection and/or serious with infection acquired in the recent or more distant past.164,206,210,298-300

Chapter 280  Toxoplasma gondii


disease.14,15,294 Non–type II strains were associated with prematurity It is also important to note that a new test has been developed that
and severity of disease at birth in a study from the United States that shows great promise for discriminating between an infection derived
used serologic tests as a method to determine the strain type.295 As from ingestion of oocysts versus tissue cysts.49 Assuming this test can
more accurate and sensitive tests for determining the genotype of an be thoroughly validated in humans, a logistically challenging task given
infecting strain are developed, this picture could change significantly. the general lack of individuals with a definitively known source of
infection, this new test could be extremely beneficial. It relies on detec-
DIAGNOSIS tion of antibodies to a protein that is abundantly expressed in sporo-
When considering toxoplasmosis in the differential diagnosis of a zoites but not in tachyzoites or bradyzoites, and it has the potential to
patient’s illness, emphasis should not be placed on whether the patient address both the epidemiologic origins of infection in different cohorts/
has been exposed to cats. Transmission of oocysts virtually always regions and could also allow a determination of whether disease initi-
occurs without the knowledge of the patient and may be unrelated to ated by one or other means leads to different clinical outcomes. What
direct exposure to a cat (e.g., transmission by contaminated vegetables, follows below concerns only the generic detection of a Toxoplasma
other foods, or water). Patients with an indoor cat that is only fed “infection,” that is, tachyzoites, without regard to the original source
cooked food are not at risk of acquiring the infection from that cat. of that infection, but such refinement is something anxiously antici-
Serologic investigation of a cat to establish whether it is a potential pated for the near future.
source of the infection should be discouraged; the prevalence of T.
gondii antibodies among cats in a given locale is usually similar to their Immunoglobulin G Antibodies
prevalence in humans. Seropositivity does not predict shedding of The most widely used tests for the measurement of IgG antibody are
oocysts. Acute toxoplasmosis has been documented in patients without the Sabin-Feldman DT,301 ELISA,302,303 the indirect fluorescent antibody
known epidemiologic risk factors for acute infection. (IFA) test,304 and the modified direct agglutination test.305 In these tests,
Because the clinical manifestations of T. gondii infection may be IgG antibodies usually appear within 1 to 2 weeks of acquisition of the
protean and nonspecific, toxoplasmosis must be carefully considered infection, peak within 1 to 2 months, fall at variable rates, and usually
in the differential diagnosis of a large variety of clinical presentations. persist for life at relatively low titers.
The correct diagnostic tests must be performed and appropriately Recent reports have shown that detecting antibodies to strain-
interpreted in light of the patient’s clinical presentation. The usefulness specific peptides can reveal the genotype of the infecting strain;
of a given diagnostic method may differ considerably with the clinical although this approach has been helpful in epidemiologic studies, it
entity, which can be toxoplasmosis in the immunocompetent or immu- has not yet been developed into a reliable, clinically useful diagnostic
nodeficient patient, ocular toxoplasmosis, toxoplasmosis in pregnancy, method.267,295
or congenital toxoplasmosis.296
Acute infection is diagnosed by characteristic serologic test results; Sabin-Feldman Dye Test
demonstration of tachyzoites in histologic sections of tissue or in cyto- The Sabin-Feldman DT is the reference serologic test against which
logic preparations of body fluids; amplification of T. gondii DNA or other methods have been evaluated.301 It is a sensitive and specific
isolation of the parasite in blood or body fluids; the demonstration of neutralization test. It measures primarily IgG antibodies that usually
a characteristic lymph node histologic appearance; or demonstration appear 1 to 2 weeks after the initiation of infection, reach peak titers
of T. gondii tissue cysts in the placenta, fetus, or neonate.8 Rarely, in 6 to 8 weeks, and then gradually decline over 6 to 12 months.8 Titers,
asymptomatic patients with latent infection have recurrent parasit- usually at low levels, probably persist for life. This test is available in
emia.297 Isolation of T. gondii from the tissues of older children or only a few reference laboratories, primarily because live organisms are
adults may only reflect the presence of tissue cysts. Finding numerous required. A negative Sabin-Feldman DT practically rules out prior
tissue cysts in tissue sections, especially associated with inflammation, exposure to T. gondii, except in patients who have been infected very
suggests but does not prove the presence of active infection. recently (e.g., within 1 to 2 weeks of exposure), are significantly immu-
nocompromised (e.g., allogeneic bone marrow transplant patients), or
Serologic Tests for Demonstration   who have a primary immunodeficiency (e.g., congenital agammaglob-
of Antibody ulinemia). Early in infection (e.g., within 4 weeks of infection), patients
The use of serologic tests for the demonstration of specific antibody to may be negative in IgG-ELISA, agglutination, or IFA kits but positive
T. gondii tachyzoites is the primary method of diagnosis. A large in the DT. However, although rare, cases of documented TE and cho-
number of tests have been described, some of which are available only rioretinitis have been reported in DT-negative patients.
in highly specialized laboratories. Different serologic tests often
measure different antibodies that possess unique patterns of rise and Indirect Fluorescent Antibody Test
fall with the time after infection.296 However, initial serologic testing The IFA test appears to measure the same antibodies as the DT, and its
can be accomplished by simultaneously requesting IgG and IgM anti- titers tend to parallel DT titers.8 False-positive results may occur with
body tests. This task can be easily achieved by hospital-based, com- sera that contain antinuclear antibodies,306 and false-negative results
mercial, or nonreference laboratories. Only positive results in IgM may occur in sera with low IgG antibody titers. Use of the Toxoplasma
antibody tests need to be sent for confirmatory testing to reference IFA test should be discouraged because of the relative high frequency
laboratories (see later). of false-negative and false-positive results.
There is no single serologic test that can be used to support the
diagnosis of acute or chronic infection by T. gondii. In most cases, a Agglutination and AC/HS Tests
battery of tests is required to enable the distinction between acute and The agglutination test using formalin-preserved whole tachyzoites is
chronic infection. Which particular combination of tests is used available commercially (bioMérieux, Marcy-l’Etoile, France) and
depends on the specific clinical category of the patient (i.e., pregnant detects IgG antibody. The test is very sensitive to IgM antibody, and
vs. immunodeficient patient; see “Clinical Manifestations”), the inter- “natural” IgM antibody causes nonspecific agglutination in sera that
val between acquisition of infection and sampling of sera,210 and the yield negative results when tested in the DT and the IFA test. This
question posed by the practitioner. The clinician must be familiar with problem is avoided by including 2-mercaptoethanol in the test. The
these problems and consult reference laboratories if the need arises. A method is accurate, simple to perform, inexpensive, and excellent for
panel of tests consisting of the Sabin-Feldman dye test (DT; detecting screening purposes.307 This method, that is, with mercaptoethanol,
IgG), the IgM-, IgA-, and IgE–enzyme-linked immunosorbent assays should not be used for the measurement of IgM antibodies.
(ELISAs), differential agglutination test (measures IgG antibody and When two different compounds (i.e., acetone and formalin) are
is also known as the AC/HS test), and the IgG avidity test is used used to fix parasites for use in the agglutination test, a “differential”

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3138
agglutination test (AC/HS test) results because the different antigenic IgG-blocking antibodies can cause false-negative results in this test
preparations vary in their ability to recognize sera obtained during the when IgG is not removed.322
acute and chronic stages of the infection.308 This test has proved useful
in helping differentiate acute from chronic infections and is best used Immunoglobulin M Enzyme-Linked
Part III  Infectious Diseases and Their Etiologic Agents

in combination with a battery of other tests. When the AC/HS yields Immunosorbent Assay
a “nonacute” pattern, the infection has been present for at least 12 The double-sandwich IgM-ELISA for detection of IgM-specific anti-
months from the time of serum sampling.309 bodies to T. gondii323-325 is currently the most widely used method for
demonstration of IgM antibodies to T. gondii in adults, the fetus, and
Immunoglobulin G Enzyme-Linked newborns.8 In contrast to the conventional method, in which the wells
Immunosorbent Assay of microtiter plates are coated with antigen, the wells are coated with
The IgG-ELISA method is now the most widely used for the demon- specific antibody to IgM. The double-sandwich IgM-ELISA is more
stration of IgG antibodies to T. gondii. Most commercial IgG antibody sensitive than the IgM-IFA test for diagnosis of recently acquired infec-
test kits are accurate for demonstration of IgG antibodies; however, it tion, and serum samples that are negative in the DT but that contain
is important to recognize that one cannot use a single IgG titer, no either antinuclear antibodies or rheumatoid factor and thus cause
matter what its level, to predict whether the infection was recently false-positive results in the IgM-IFA test are negative in the double-
acquired or acquired in the distant past. sandwich IgM-ELISA. This latter observation is attributed to the fact
that serum IgM fractions are separated from IgG fractions during the
Immunoglobulin G Avidity Test initial step in the double-sandwich IgM-ELISA procedure.8
A number of tests for avidity of Toxoplasma IgG antibodies have been Despite the wide distribution of commercial test kits to measure
introduced to help differentiate between recently acquired and distant IgM antibodies, these kits often have low specificity, and the reported
infection.310 This method is based on the observation that during acute results are frequently misinterpreted. False-positive results and the
T. gondii infection, IgG antibodies bind antigen weakly (i.e., have low problems associated with the persistence of positive titers, even years
avidity), whereas chronically infected patients have more strongly after the initial infection, remain major obstacles to correct interpreta-
binding (high avidity) antibodies.310 Protein-denaturing reagents, tion of the results obtained in these tests.316
including urea, are used to dissociate the antibody-antigen complex.
Low or equivocal avidity test results can persist for months to years Immunoglobulin M Immunosorbent
after the primary infection,310 and for this reason, a low or equivocal Agglutination Assay
avidity test result must not be used to determine whether the infection The IgM immunosorbent agglutination assay (IgM-ISAGA) (bioMéri-
was acquired recently. The time of conversion from low or equivocal eux), which binds the patient’s IgM to a solid surface and uses intact,
to high avidity is highly variable among different individuals, including killed tachyzoites to detect IgM antibodies, is highly sensitive.326 The
pregnant women.298,311,312 However, it has been demonstrated that once test is simple to perform, does not require the use of enzyme conjugate,
the avidity test result is high, the patient was infected at least 3 to 5 and is read in the same manner as the agglutination test. Overall, it is
months earlier.312 This timing depends on the method used. High- more sensitive and specific than the IgM-IFA test. The presence of
avidity test results by the IgG-VIDAS avidity test (VIDAS Toxoplasma rheumatoid factor or antinuclear antibodies does not cause false-
IgG Avidity, bioMérieux), for example, have been essentially found positive results in the IgM-ISAGA. In adults, it is more sensitive but
only in pregnant women who have been infected for at least 4 much less specific than the double-sandwich IgM-ELISA method. In
months.300,313,314 infants the IgM-ISAGA is the most sensitive method and is used effec-
The avidity test should only be used as an additional confirmatory tively for diagnosis of congenital infection in infants 6 months of age
diagnostic method in patients with positive and/or equivocal IgM test or younger.327 A positive IgM-ISAGA test result in the first 10 days of
results or when the results of a battery of tests are equivocal or inter- life should be repeated after 10 days to rule out the possibility of mater-
preted as consistent with the possibility of a recently acquired infec- nal contamination of IgM antibodies. The ISAGA method has also
tion. Health care providers involved in the care of pregnant women been used to detect IgA and IgE antibodies.
should be aware that avidity testing is only a confirmatory test. It
should not be used alone as a definitive test for decision making. Immunoglobulin A Antibodies
IgA antibodies may be detected in sera of acutely infected adults and
Immunoglobulin M Antibodies congenitally infected infants using ELISA or ISAGA.328-330 As is true
IgM antibodies may appear earlier and decline more rapidly than IgG for IgM antibodies to the parasite, IgA antibodies may persist for many
antibodies. IgM antibody tests have been widely used for the diagnosis months or more than 1 year. However, IgA antibodies tend to disap-
of acute infection and to determine whether a pregnant woman has pear earlier than the IgM, and their presence at high titers is usually
been infected during gestation or before conception. There has been a associated with early infections (e.g., within 3 months of the date of
heightened awareness of the fact that titers in tests for IgM antibodies serum sampling). The increased sensitivity of IgA assays over IgM
may persist for years after the acute infection and that the reliability of assays for the diagnosis of congenital toxoplasmosis represents a major
commercially available assays varies considerably.315-317 Both the labo- advance in the serologic diagnosis of the infection in the fetus and
ratory performing the test and the physician requesting the test should newborn.330 IgA antibodies are rarely detectable by ELISA in sera of
be aware of this problem. In 1997, the U.S. Food and Drug Administra- AIDS patients with TE.330 If IgA antibodies are detected in the newborn
tion (FDA) issued a health advisory warning about the use of T. gondii during the first 10 days of life, the test should be repeated 10 days after
IgM commercial test kits as the sole determinant of recent infection in birth to make certain that what is being measured is not contaminating
pregnant women.318 At present, the decision to treat or undertake other maternal IgA antibodies. The possibility that such contamination
medical interventions, including the termination of pregnancy, should might occur is the reason that under most circumstances, peripheral
be based on clinical evaluation and additional testing performed in blood rather than cord serum be used to measure IgM, IgA, or IgE
reference or research laboratories with experience in the diagnosis of antibodies in the newborn.
toxoplasmosis. For further discussion, see “Toxoplasma gondii Infec-
tion in Pregnancy.”319 The persistence of IgM antibodies for several Immunoglobulin E Antibodies
years appears not to have clinical significance.8 IgE antibodies are detectable by ELISA in sera of acutely infected
adults,331,332 congenitally infected infants,331,332 and children with con-
Immunoglobulin M Indirect Fluorescent genital toxoplasmic chorioretinitis.333 The duration of IgE seropositiv-
Antibody Test ity is briefer than that with IgM or IgA antibodies and hence appears
IgM-IFA antibody appears within the first week of infection; titers useful for identifying recently acquired infections.210,332 T. gondii–
rise rapidly and then fall to low titers and usually disappear within a specific IgE antibody has been detected in patients with TE and may
few months. Low titers may persist 1 year or longer.320 Antinuclear be useful as a marker for TE in this population of patients.332 IgE anti-
antibodies and rheumatoid factor may cause false-positive results.321 bodies have been reported to be present in 85.7% of asymptomatic

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3139
29
seroconverters and in 100% of seroconverters with overt toxoplasmo- CNS of patients with TE. Both the fluorescent antibody and immu-
sis.334 For neonatal diagnosis of congenital toxoplasmosis, IgE was less noperoxidase methods are applicable to unfixed or formalin-fixed
sensitive than IgM and IgA, but simultaneous measurement of the paraffin-embedded tissue sections.29 Fluorescein-labeled monoclonal
three immunoglobulins at birth improved the diagnostic yield to antibodies to T. gondii for staining touch preparations of specimens353

Chapter 280  Toxoplasma gondii


81%.334 Emergence of specific IgE during postnatal treatment for con- and rapid electron microscopy354 have been used successfully to diag-
genital toxoplasmosis may indicate poor adherence or inadequate nose TE.
dosing. A rapid, technically simple, but underused method is the detection
of T. gondii in air-dried, Wright-Giemsa–stained slides of centrifuged
Polymerase Chain Reaction (e.g., cytocentrifuged) sediment of CSF or of brain aspirate or in
PCR amplification for the detection of T. gondii DNA in body fluids impression smears of biopsy tissue.
and tissues has successfully diagnosed congenital,279,335 ocular,178,179 Endomyocardial biopsy has been used successfully to diagnose
pulmonary,176 cerebral, and disseminated toxoplasmosis.336,337 PCR of toxoplasmosis in heart transplant recipients.355 Characteristic histo-
amniotic fluid has revolutionized the diagnosis of intrauterine T. gondii logic criteria alone are probably sufficient to establish the diagnosis
infection by enabling an early diagnosis to be made, thereby avoiding of TL in older children and adults (see “Lymph Node” under
the use of invasive procedures on the fetus.8,335 PCR has also been suc- “Pathology”).163
cessfully used on samples of CSF, blood, urine, and placental and fetal
tissues for diagnosis of congenital infection.8,338-341 The sensitivity of Isolation of Toxoplasma gondii
PCR in CSF varies between 11% and 77%, whereas the specificity is Although largely replaced by use of PCR, isolation of T. gondii from
close to 100%.336 PCR may also detect the parasite in buffy coat speci- blood or body fluids can also be used to establish that the infection is
mens of AIDS patients with TE.337 The sensitivity of PCR on whole acute. In neonates, isolation of the organism from the placenta is highly
blood or buffy coat ranges from 15% to 85%. PCR on blood appears suggestive of fetal involvement, and isolation from fetal tissues is diag-
to be a valuable tool primarily in patients with disseminated disease; nostic of congenital infection.8 Attempts at isolation of the parasite can
it is less sensitive in the detection of TE because a relatively low per- be performed by mouse inoculation or inoculation of tissue cell cul-
centage of AIDS patients with TE have parasitemia.342,343 Therapy for tures.8 In tissue cell cultures, parasite-laden cells can be demonstrated
toxoplasmosis appears to influence the sensitivity of the method; sen- with appropriate staining, and plaques are formed in which tachyzoites
sitivity is higher in CSF or blood samples collected before or within are easily recognized. Tissue cell culture has the advantage of wide-
the first week of therapy.337 In a recent case report, quantitative real- spread availability (e.g., virology laboratories) and yields results more
time PCR proved useful for the diagnosis and monitoring of Toxo- rapidly (within 3 to 6 days) than does mouse inoculation.
plasma infection in a heart transplant recipient who developed
toxoplasmosis after TMP-SMX for PCP prophylaxis had been stopped. Radiologic Methods
Decreasing parasitic burdens in sequential samples of cerebrospinal Radiologic studies are of particular help in patients with toxoplasmosis
fluid, blood, and BAL fluid correlated with a favorable outcome and of the CNS. The presence of calcifications in the brain of a newborn,
allowed modulation of the immunosuppressive drug regimen in this detected by radiography, ultrasonography, or CT, should heighten the
patient.344 Peripheral blood PCR has also been found to be positive in suspicion of T. gondii as the cause of the disease (Fig. 280-6A). In
immunocompetent patients in Brazil who present with symptomatic severely affected infants with congenital toxoplasmosis, unilateral or,
ocular toxoplasmosis as a result of reactivation of an infection acquired more often, bilateral and symmetrical dilatation of the ventricles is a
in the distant past.345 It is unclear whether these findings only apply to common finding.8
patients in this area of the world, where atypical and more virulent In the majority of immunodeficient patients with TE, CT scans
strains are common in contrast to Europe and the United States. show multiple bilateral cerebral lesions.356 Although multiple lesions
Because there is no standardized PCR assay, performance charac- are more common in toxoplasmosis, they also may be solitary; a single
teristics will vary widely depending on the laboratory, gene target, lesion should not exclude TE as a diagnostic possibility. Clinicians
primers, and sample preparation.346 Primers targeting multicopy B1 or should be aware that toxoplasmosis may manifest as an encephalitis
AF146527 (also known as the 529-bp repeat element) genes appear to that at autopsy is “diffuse,” in which case, the neuroimaging study
be the most sensitive and are the most broadly used.346-348 A number results may appear normal or reveal findings suggestive of HIV
of investigators have reported a higher sensitivity of the 529-bp target encephalopathy.174
for the prenatal diagnosis of congenital toxoplasmosis when compared CT scans in AIDS patients with TE reveal multiple ring-enhancing
with the B1 gene.349 For maximal reliability, clinical samples should lesions in 70% to 80% of the cases.236 In AIDS patients who are not
be sent to reference laboratories experienced in performing this receiving appropriate ART or anti-Toxoplasma prophylaxis but have
assay.340,350 detectable Toxoplasma IgG and multiple ring-enhancing lesions seen
A retrospective analysis of French laboratories performing routine on CT or MRI, the predictive value for TE is approximately 80%.357
surveillance on blood from HSCT patients using PCR for the AF146527 Lesions tend to occur at the corticomedullary junction (frequently
gene found that 23 of 1220 blood samples (1.9%) were positive, a lower involving the basal ganglia) and are characteristically hypodense.358,359
figure than other series.350a Fifteen patients were not receiving TMP- The number of lesions is frequently underestimated by CT, although
SMX prophylaxis and were treated for toxoplasmosis. The other delayed imaging after a double dose of intravenous (IV) contrast mate-
eight were simply continued on TMP-SMX prophylaxis. No clinical rial may improve the sensitivity of this modality.358-360 An enlarging
signs of toxoplasmosis resulted. This and other studies suggest that hypodense lesion that does not enhance is a poor prognostic sign.361
a PCR-based pre-emptive strategy is useful in the earlier diagnosis TE lesions on MRI studies appear as high-signal abnormalities on
and treatment of Toxoplasma infection, before patients develop organ T2-weighted studies and reveal a rim of enhancement surrounding the
involvement.232,350b edema on T1-weighted contrast-enhanced images (see Fig. 280-6B and
C). MRI has superior sensitivity compared with CT, particularly if
Histologic Diagnosis gadolinium is used for contrast, and often demonstrates a lesion or
Demonstration of tachyzoites in tissue sections or smears of body fluid lesions or more extensive disease not seen by CT.360,362 Hence, MRI
(e.g., CSF, amniotic fluid, or BAL) establishes the diagnosis of acute should be used as the initial procedure when feasible (or if a single
infection or reactivation of a latent infection.8 It is often difficult to lesion is demonstrated by CT). Nevertheless, even characteristic lesions
demonstrate tachyzoites in stained tissue sections. Multiple tissue cysts on CT or MRI studies are not pathognomonic of TE. The major dif-
near an inflammatory necrotic lesion can be considered as diagnostic ferential diagnosis of focal CNS lesions in AIDS patients is CNS lym-
of acute infection or reactivation of a latent infection.351 Fluorescent phoma, which may manifest with multiple enhancing lesions in 40%
antibody staining may be useful, but this method often yields nonspe- of cases. The probability of TE falls and the probability of lymphoma
cific results.352 The immunoperoxidase technique, which uses antisera rises in the presence of single lesions on MRI.356 A brain biopsy may
to T. gondii, has proved both sensitive and specific; it has been used therefore be required in the patient with a solitary lesion, especially if
successfully in clinical settings to demonstrate the organisms in the confirmed by MRI, to obtain a definitive diagnosis.363 In non-AIDS

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Part III  Infectious Diseases and Their Etiologic Agents 3140

C
FIGURE 280-6  Imaging studies of central nervous system toxoplasmosis. A, Computed tomography (CT) scan of an infant born with congenital
toxoplasmosis, illustrating the calcifications and hydrocephalus that are typically seen in the brain. B, CT scan with contrast enhancement (left) and
T2-weighted magnetic resonance imaging (MRI) scan (right) of an acquired immumodeficiency syndrome (AIDS) patient with toxoplasmic encephalitis,
demonstrating multiple lesions, which are more easily identified in the MRI scan. C, T1-weighted MRI scan without (left) and with (right) contrast enhance-
ment, of an AIDS patient with toxoplasmic encephalitis. Note the ring-enhancing lesion on the right.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3141
immunocompromised patients, TE can also present as single or mul- In the presence of clinical illness, it is important to establish whether
tiple ring-enhancing lesions. However, additional etiologies, including the patient’s symptoms are due to a recently acquired infection or to
invasive fungal, nocardial, and mycobacterial infections, are more recrudescence of latent infection (chronic infection) or are unrelated
common in these patients with this radiologic presentation than in to the infection. A true negative IgM test in an otherwise immunologi-

Chapter 280  Toxoplasma gondii


AIDS patients. Early brain biopsy and empirical treatment with a wider cally normal individual essentially rules out that the infection has been
antimicrobial spectrum is usually required in non-AIDS immunocom- acquired in recent months. A positive IgM test is more difficult to
promised patients with multiple space-occupying brain lesions. interpret correctly. One must not assume that a positive IgM test result
In AIDS patients with TE, CT-scan improvement is seen in up to is diagnostic of recently acquired infection. The presence of T. gondii–
90% of patients after 2 to 3 weeks of treatment.356,358 Complete resolu- specific IgM antibodies can be interpreted as a true-positive result
tion takes from 6 weeks to 12 months; peripheral lesions resolve more consistent with recently acquired infection, a true-positive result con-
rapidly than deeper ones. Smaller lesions usually resolve completely on sistent with a chronic infection (IgM antibodies have been shown to
MRI studies within 3 to 5 weeks, but lesions with a mass effect tend to persist in some individuals for as long as several years after the acute
resolve more slowly and leave a small residual lesion.364 A radiologic infection), or a false-positive result. To establish which of these is most
response to therapy lags behind the clinical response, with better cor- likely in a given case, confirmatory testing in a reference laboratory
relation between them observed by the end of acute therapy.365 should be performed whenever feasible.316,318
CT and MRI in toxoplasmic myelopathy usually demonstrates The use of a combination or battery of tests has been useful for
localized enlargement of the spinal cord,239,366 which may result in determining whether the patient has been recently infected or infected
obstruction to dye flow on myelography.239 Gadolinium enhancement in the more distant past.210,299 If the patient has received a blood
of MRI studies usually highlights as an intramedullary lesion at the site transfusion, serologic tests may measure exogenously administered
of spinal cord enlargement.366 rather than endogenous antibody. The use of serologic tests to evalu-
A variety of positron-emission tomography (PET) scanning,367 ate the response to therapy should be discouraged. The PAMF-TSL
radionuclide scanning,368 and MRI techniques369 have been used currently serves as a reference laboratory. Physicians in the PAMF-
to evaluate AIDS patients with focal CNS lesions, specifically to differ- TSL also offer interpretation of test results and consultation on treat-
entiate between toxoplasmosis and primary CNS lymphoma.369 ment and patient management to clinicians in the United States and
18
F-fluorodeoxyglucose PET scanning is now widely used in the evalu- worldwide.
ation of patients with tumors. There is a significantly higher uptake of
18
F-fluorodeoxyglucose in patients with cerebral lymphoma than in Toxoplasmosis in the
patients with TE.370 Radionuclide scanning has also been used to dif- Immunocompetent Patient
ferentiate between CNS toxoplasmosis and lymphoma. Neoplasms Tests for IgG and IgM antibodies should be used for initial evaluation
usually demonstrate increased uptake of thallium-201 on both early and of immunocompetent patients. Testing of serial specimens obtained 3
late scanning.369 Although published studies suggest a high sensitivity weeks apart (in parallel) provides the best discriminatory power if the
and specificity of these studies, in practice they often are not helpful, in results in the initial specimen are equivocal. Negative results in both
part because of variability in uptake and in part because they are often of these tests virtually rules out the diagnosis of toxoplasmosis. Early
used after an empirical trial of anti-Toxoplasma therapy. Thallium scans in infection, IgG antibodies may not be detectable, whereas IgM anti-
may have decreased diagnostic utility in the setting of ART.371 bodies are present, hence the need for both tests to be performed.
Proton magnetic resonance spectroscopy to evaluate brain lesions Acute infection is supported by documented seroconversion of IgG or
has been used in a few patients. Magnetic resonance spectroscopy in IgM antibodies or a greater than two-tube rise in antibody titer in sera
patients with TE reveals an elevation in the lactate and lipid contents369 run in parallel. A single high titer of any immunoglobulin antibodies
and a decrease in the levels of choline. In contrast, MR spectroscopy is insufficient to make the diagnosis; IgG antibodies may persist at high
in patients with CNS lymphoma reveals mildly elevated levels of titers for many years,8 and IgM antibodies may be detectable for more
choline.369 than 12 months. When only a single serum sample is available, a
battery or combination of tests is usually required in determining the
Cerebrospinal Fluid Abnormalities likelihood that the infection is acute.
CSF abnormalities in patients with TE are nonspecific; mild mono- Toxoplasmosis should be considered in the differential diagnosis of
nuclear pleocytosis and mild to moderate elevations in CSF protein are lymphadenopathy, whether or not symptoms are present and especially
often observed, and hypoglycorrhachia is uncommon.198,372 Almost in those without symptoms. Confirmatory serologic tests should be
unique to infants with neonatal toxoplasmosis, however, is the very obtained in such patients. The interval between the clinical onset of
high protein content of the ventricular fluid and eosinophilia. Although, lymphadenopathy and the date that the specimen is drawn is critical
in some infants, the protein level is just slightly above normal, in others for interpretation of the test results.210 In patients whose serum is
it can be measured in grams per deciliter rather than in milligrams per available during the first 3 months after the clinical onset, at least
deciliter.8 Demonstration of intrathecal production of T. gondii–specific the IgG test and the IgM-ELISA are positive. In those patients in
IgG or IgM in the absence of CSF contamination with blood is diag- whom sera are obtained more than 3 months after the clinical onset,
nostic of TE.373,374 the IgM-ELISA is most likely to be negative, but the IgG test and at
least one of the following tests are positive: IgA-ELISA, IgE-ELISA,
Diagnosis of Specific Clinical Entities IgE-ISAGA, or AC/HS test.210 A high IgG avidity test result in an
The initial step in pursuing the diagnosis of T. gondii infection or individual who has recent onset of lymphadenopathy (e.g., within 2 to
toxoplasmosis is to determine whether the patient has been exposed 3 months of sera sampling) suggests a cause other than toxoplasmo-
to the parasite. In virtually all cases, tests for IgG antibodies reliably sis,309 and further workup is warranted. A nonacute AC/HS pattern
establish the presence or absence of the infection; a negative IgG test in an individual at less than 12 months after clinical onset of lymph-
essentially rules out prior or recent exposure to the parasite. However, adenopathy should suggest an etiology other than TL. In such cases,
clinicians should be aware that IgG antibodies may be absent in immu- investigation for alternative causes, including malignancy, should be
nocompetent patients who have been tested within the first 2 weeks of undertaken.309
the acute infection, in patients with severe immunodeficiencies involv- Histologic diagnosis can be useful in some cases of suspected toxo-
ing defective production of IgG antibodies, and in BMT patients plasmosis in the immunocompetent patient. The histologic criteria for
despite having had detectable IgG antibodies in the pretransplantation the diagnosis of TL has been well established (see Fig. 280-3A) (see
period. Because of the higher sensitivity of the IgG DT, on occasion, “Histologic Diagnosis”).163 In this setting, there is no need to visualize
IgG antibodies are initially negative in commercially available kits but the parasite. Endomyocardial biopsy and biopsy of skeletal muscle have
positive in the IgG DT (see “Serologic Tests for Demonstration of been successfully used to establish T. gondii as the etiologic agent of
Antibody”). In addition, cases of documented toxoplasmic chorioreti- myocarditis and polymyositis in rare cases in immunocompetent
nitis and TE in adult patients have been observed in which IgG anti- patients.164 Isolation studies and PCR have rarely proved useful in
bodies were not demonstrable; such cases are very uncommon. immunocompetent patients.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3142
Toxoplasmosis in the   tissue cyst may only reflect chronic infection unless it is associated with
Immunodeficient Patient an area of inflammation (e.g., as seen in myocardial biopsy); however,
Because reactivation of chronic infection is the most common cause visualization of several tissue cysts virtually always means that active
of toxoplasmosis in patients with AIDS, malignancies, or organ trans- infection is present.
Part III  Infectious Diseases and Their Etiologic Agents

plants, initial assessment of these patients should routinely include an When clinical signs suggest involvement of the CNS or spinal cord,
assay for T. gondii IgG antibodies. IgG antibody testing should be the workup should include CT or MRI (see “Radiologic Methods”) of
ideally performed as soon as it is established that the patient is immu- the brain. These studies should be performed even if the neurologic
nocompromised or is about to be immunosuppressed. Those with a examination does not reveal focal deficits.
positive result are at risk of reactivation of the infection; those with a A lumbar puncture should be performed if it can be done safely,
negative result should be instructed on how they can prevent becoming ideally before or soon after initiation of therapy; PCR can then
infected (see “Prevention”). Those at highest risk (e.g., HIV-infected be performed on the CSF specimen. A positive Toxoplasma PCR essen-
patients not receiving appropriate ART or anti-Toxoplasma prophy- tially confirms the diagnosis of TE. CSF can also be sent for isolation
laxis) who are initially seronegative should be retested on an annual studies if available. PCR examination of the CSF also can be used for
basis to determine whether they have seroconverted. Sero­negative the detection of Epstein-Barr virus, JC virus, or CMV DNA in patients
organ transplant recipients should be identified before transplantation in whom primary CNS lymphoma, progressive multifocal leukoen-
because they will be at risk for infection if a seropositive donor who cephalopathy, or CMV ventriculitis, respectively, have been entertained
can potentially transmit the parasite via the allograft is selected. In this in the differential diagnosis. Especially in HIV-infected patients, a posi-
setting, administration of anti-Toxoplasma prophylaxis in the post- tive Epstein-Barr virus PCR in the setting of a focal contrast-enhancing
transplantation period can avoid unnecessary morbidity and CNS lesion is strongly suggestive of CNS lymphoma.377 Demonstration
mortality.81 of the intrathecal production of T. gondii–specific antibody within the
In patients with AIDS and toxoplasmosis, the IgG titer may be CSF may help confirm the diagnosis of TE.374 Unless sufficient CSF is
relatively low, and tests for IgM, IgA, and IgE antibodies are usually available, we suggest that the highest priority be for PCR and an attempt
negative.236 at isolation of the parasite. Detection of T. gondii–specific IgM has a
In the early postoperative period in heart transplant recipients with very low yield and is not recommended.
pretransplantation Toxoplasma antibodies who present with a clinical Empirical anti–T. gondii therapy for patients with multiple ring-
illness, serologic test results may be misleading.221,355 In these patients, enhancing brain lesions (usually established by MRI), positive IgG
results indicating apparent reactivation (a rising IgG and IgM titer) antibody titers against T. gondii, and AIDS (e.g., patients with a CD4
may be present in the absence of clinically apparent infection. In addi- count of <200 cells/mm3) is accepted practice; a clinical and radiologic
tion, serologic test results consistent with chronic infection may be response to specific anti–T. gondii therapy essentially confirms the
seen in the presence of toxoplasmosis.221,355 In heart transplant recipi- diagnosis of TE (Fig. 280-7). Use of adjunctive corticosteroids to
ents in whom toxoplasmosis is suspected as a cause of altered myocar- decrease cerebral edema can cause temporary clinical improvement
dial function, endomyocardial biopsy has proved useful.355 The parasite that is incorrectly attributed to the pyrimethamine and sulfa. This
has been demonstrated in the myocardium of patients in whom the empirical approach is not recommended for non-AIDS immunocom-
biopsy was performed because of a suspicion of rejection.355 PCR promised patients with this presentation because the differential diag-
testing of peripheral blood, BAL, CSF, vitreous fluid, and other body nosis is wider and often includes other etiologic agents, such as invasive
fluids or tissues may prove to be useful in patients with solid-organ molds, nocardia, bacterial brain abscess, and mycobacteria. In these
transplants who are suspected to have toxoplasmosis. patients, an early brain biopsy is recommended. Brain biopsy should
Diagnosis in the bone marrow transplant recipient often requires be considered in AIDS patients with presumed TE if there is a single
special consideration. It is critical in all BMT patients that a serum IgG lesion on MRI, a negative IgG antibody test, an inadequate clinical
titer be performed before the transplantation. Toxoplasmosis in these response (within a 2- to 3-week period) or progression during optimal
patients is almost always due to recrudescence of a latent infection. therapy, or in patients whom the physician considers to have adhered
After BMT, the preexisting IgG antibody titer may rise, remain stable, to an effective prophylactic regimen against T. gondii (e.g., TMP-SMX).
decrease, or become negative. Thus, post-transplantation serology fre- An impression smear of the brain biopsy specimen can be made and
quently is not helpful in this group of patients and emphasizes the immediately examined for the presence of tachyzoites by using the
need for knowing the patients’ pretransplantation serologic status. conventional Wright-Giemsa stain used for blood smears in most labo-
Clinical evidence of encephalitis, pneumonia, fever, brain lesions, or ratories. The brain specimen should then be submitted to the pathol-
any other unexplained syndrome in BMT patients with preexisting T. ogy and microbiology departments for appropriate workup. In addition
gondii IgG antibodies must include toxoplasmosis in the differential to hematoxylin and eosin staining, T. gondii–specific immunoperoxi-
diagnosis. The ultimate diagnosis of toxoplasmosis in these patients dase staining should be performed. Because the amount of brain tissue
requires the use of histologic, PCR, or isolation methods to detect the obtained at aspiration or biopsy is usually small, sufficient tissue for
presence of the parasite. In patients with allogeneic HSCT who are IgG mouse inoculation may not be available; however, this should be per-
antibody test positive for T. gondii before transplantation, routine PCR formed whenever feasible. A positive result may often be obtained with
testing of peripheral blood specimens in the post-transplantation far less than 1 g of brain tissue. PCR has been used successfully in brain
period has been proposed as an appropriate tool for guiding preemp- tissue to diagnose TE,378 but a positive result should be interpreted with
tive therapy. In one report of 106 patients, toxoplasmosis developed caution because it may not distinguish between the patient with TE
in 38% of those who were PCR positive versus 0% in those who were from one with latent infection (asymptomatic carrier of brain tissue
PCR negative.232 In later studies, however, a first positive PCR was seen cysts) who has CNS pathology resulting from a process other than
only at the time of presentation with clinical symptoms or subsequent toxoplasmosis.
to that.350b,375 In the appropriate clinical setting, it is important to include toxo-
Serologic tests in patients with hypoglobulinemia or agammaglob- plasmosis in the differential diagnosis of pulmonary symptoms, par-
ulinemia may not be useful to diagnose toxoplasmosis; active infection ticularly in those individuals with interstitial infiltrates or ground-glass
can occur in these patients in the setting of negative IgG titers. opacities. Wright-Giemsa staining and PCR of BAL specimens are
A definitive diagnosis of toxoplasmosis in the immunodeficient useful for the diagnosis of pulmonary toxoplasmosis.176,379
patient relies on histologic demonstration of the parasite (usually in In patients with visual symptoms in whom toxoplasmic chorioreti-
association with an inflammatory process), on detection of T. gondii nitis is a possibility, PCR examination of vitreous or aqueous fluid can
DNA by PCR, or on isolation of the parasite. PCR or attempts to isolate be considered and is particularly helpful in patients with atypical clini-
the parasite can be performed in essentially any body fluid or tissue cal features of toxoplasmic chorioretinitis and in immunocompro-
that is clinically affected; peripheral blood testing by PCR (and in some mised patients.179,266,380 Of note, PCR examination of the vitreous fluid
instances for isolation) should be considered in immunocompromised can also be helpful when other etiologic agents, such as herpes simplex
patients suspected to have toxoplasmosis. The presence of tachyzoites virus, varicella-zoster virus, or CMV, are considered in the differential
is diagnostic of active infection. The presence of a solitary T. gondii diagnosis.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3143
CNS signs/symptoms in HIV-positive patients

Chapter 280  Toxoplasma gondii


Unknown Toxoplasma lgG antibody Negative Diagnosis of TE
unlikely

Positive
Request serology and proceed
with algorithm that favors
empirical treatment until
results are known
CT/MRI with
contrast (MRI
preferable)

No lesions
Single lesion (if CT is negative,
MRI is advised) Workup for causes
other than TE.
MRI if CT was Multiple lesions Repeat MRI in 42-78
If MRI not
initially performed on CT/MRI hours if all
available
investigations negative.

Single lesion Multiple lesions

Long list of possibilities if other Presumptive diagnosis


symptoms, signs, or laboratory Improved of TE. Continue
investigations are unrevealing Brain biopsy Treat empirically clinically treatment for 3-6
not available for TE by 7 days weeks, followed by
maintenance therapy.

Consider lumbar puncture (if the risk No improvement


for herniation is judged to be low) clinically by 10-14 days
PCR for: or clinical deterioration by day 3
T. gondii
Epstein-Barr virus
JC virus
Cytology
Consider brain biopsy
Isolation, PCR
Histopathology Definitive diagnosis
Immunoperoxidase
AFB, Giemsa, Gram stains

FIGURE 280-7  Diagnostic approach and management algorithm for human immunodeficiency virus (HIV)-infected patients with central
nervous system (CNS) symptoms or signs that might potentially be toxoplasmic encephalitis (TE). AFB, acid-fast bacilli; CT, computed tomog-
raphy; IgG, immunoglobulin G; MRI, magnetic resonance imaging; PCR, polymerase chain reaction.

The intraocular production of T. gondii–specific IgG antibodies has In pursuing the diagnosis, histologic examination with the appro-
also been reported to be diagnostically useful.381 Local antibody pro- priate stains and mouse inoculation can be attempted in virtually any
duction in aqueous humor can be quantified by calculating the tissue suspected of being involved by T. gondii. Body fluids that should
Goldmann-Witmer (GW) coefficient. The GW coefficient expresses be considered for examination by PCR include CSF, blood, vitreous,
the level of Toxoplasma-specific IgG relative to the level of total IgG in aqueous, and BAL specimens. Reference laboratories should be con-
the aqueous humor as a fraction of the level of Toxoplasma-specific tacted before diagnostic procedures to optimize the handling of the
IgG relative to the level of the total IgG in the serum.382 The GW coef- specimens and their yield.
ficient = Toxoplasma IgG/total IgG in aqueous humor ÷ Toxoplasma
IgG/total IgG in serum. A coefficient greater than 2 is considered posi- Ocular Toxoplasmosis
tive and diagnostic of ocular toxoplasmosis. It has been well established In patients with active chorioretinitis caused by reactivation of con-
that, in contrast to ocular viral infections, the GW coefficient is a more genital T. gondii infection, low titers of IgG antibody are usual and IgM
sensitive method than PCR for the diagnosis of ocular toxoplasmosis. antibodies are not usually detected. In contrast, in patients with cho-
However, PCR in vitreous fluid appears to be superior to the GW coef- rioretinitis as a result of an acute infection, IgG and IgM antibodies
ficient in immunocompromised patients, the elderly, and those with will be detected. Positive IgM test results should always be confirmed
atypical lesions and extensive retinochoroiditis. at a reference laboratory.382
PCR and isolation studies in peripheral blood can help establish T. In most cases, toxoplasmic chorioretinitis is diagnosed by oph-
gondii as the etiologic agent of a febrile syndrome or systemic symp- thalmologic examination, and empirical therapy directed against the
toms of unclear cause.337,383 These studies tend to have a higher yield organism is often instituted based on clinical findings and serologic
early in the disease and before or shortly after specific anti–T. gondii test results. In a number of patients, the morphology of the retinal
therapy is initiated.337 lesion or lesions may be nondiagnostic, or the response to treatment

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3144
may be suboptimal, or both. In such cases (unclear clinical diagnosis battery of other tests, in women in their first 16 weeks of gestation,
or inadequate clinical response, or both), the detection of an abnor- detection of high-avidity IgG antibodies by the IgG-VIDAS avidity
mal T. gondii–antibody response in ocular fluids (Goldman-Witmer test, for example, can be a useful addition to the discriminatory
coefficient),380,384 or demonstration of the parasite by isolation, histo- power of a combination of tests in distinguishing recently acquired
Part III  Infectious Diseases and Their Etiologic Agents

pathologic examination, or PCR have been used successfully to estab- from chronic infection.296,300,314 It is critical to recognize that the value
lish the diagnosis.382,385 PCR has been used in both vitreous and of the avidity test is in the first 3 to 5 months of gestation (i.e., the
aqueous fluids in an attempt to support or confirm the diagnosis of T. fetus of a woman in the 14th week of gestation with a positive IgM
gondii as the cause of the retinal lesions.178,179,266,380 In patients in test and a high-avidity result is not at risk for congenital toxoplasmo-
whom toxoplas­mosis is considered in the differential diagnosis but in sis).300 Because low or equivocal avidity test results may persist for
whom the presentation is atypical, PCR is a useful diagnostic aid. many months, their presence does not necessarily indicate recently
Vitreous biopsy is a potentially hazardous procedure, and its use acquired infection.
should be considered only when other diagnostic measures have not Confirmatory testing, using a battery of tests and the VIDAS avidity
revealed a cause. method in pregnant women during their first 24 weeks of gestation,
has the potential to decrease the need for follow-up sera and thereby
Toxoplasma gondii Infection in Pregnancy reduce costs, to make the need for PCR on amniotic fluid and for treat-
Acute acquired T. gondii infection is diagnosed serologically by the ment of the mother with spiramycin unnecessary, to remove the preg-
same methods used for immunocompetent adults discussed earlier nant woman’s anxiety associated with further testing, and to decrease
(Fig. 280-8).319 Special care is taken to determine whether the infection unnecessary abortions.298-300 Although the avidity test represents an
was acquired before or after conception. This determination is fre- additional confirmatory method (most useful if high-avidity antibod-
quently difficult because routine serologic screening is not conducted ies are detected within the first 16 weeks of gestation), it should not be
in pregnant women in the United States. used as the only confirmatory test for pregnant women with positive
The diagnosis of acute T. gondii infection or toxoplasmosis ideally IgG and/or IgM antibodies because of the potential to misinterpret
requires demonstration of a rise in titers in serial serum samples (either low- or borderline-avidity antibody results.
conversion from a negative to a positive titer or a fourfold rise from a Once the diagnosis of acute acquired infection during pregnancy
low to a higher titer).8 These specimens should be obtained at least 3 has been presumptively established, diagnostic efforts should focus on
weeks apart and be tested in parallel. Because the diagnosis is fre- determining whether the fetus has been infected.
quently considered relatively late in the course of the patient’s preg-
nancy, serologic test titers may already have reached their peak at the Congenital Infection in the Fetus  
time the first serum is obtained for testing. Therefore, it is often difficult and Newborn
to discriminate between infections acquired recently (possibly during Prenatal diagnosis of fetal infection is advised when a diagnosis of
pregnancy) and those acquired in the more distant past, when the only acute infection is established or highly suspected in a pregnant woman.
available serum sample has been obtained in the third trimester of the Methods to obtain fetal blood, such as periumbilical fetal blood sam-
pregnancy. Thus, the initial serum should be obtained as early as pos- pling, have been largely abandoned because of the rate of false-negative
sible during gestation. prenatal diagnoses, the risk involved for the fetus, and the delay in
Initial screening of maternal serum involves testing for IgG and obtaining definitive results with conventional parasitologic tests.279
IgM antibodies; a lack of both immunoglobulin antibodies essentially Prenatal diagnosis of congenital toxoplasmosis is presently based
excludes active infection but identifies the patient as being at risk for on ultrasonography and amniocentesis. PCR on amniotic fluid for the
acquisition of the infection and hence in need of instruction about detection of T. gondii–specific DNA performed at 18 weeks of gesta-
primary prevention. The presence of IgG antibodies in the absence of tion or later is more sensitive, more rapid, and safer than conventional
IgM antibodies in the first two trimesters almost always indicates diagnostic procedures involving fetal blood sampling.279,387 Amniotic
chronic maternal infection with essentially no risk to the fetus (the fluid should be tested by PCR in all cases, with serologic test results
exceptions are severely immunodeficient patients). In the third trimes- diagnostic of or highly suggestive of acute acquired infection during
ter, a negative IgM test titer is most likely consistent with a chronic pregnancy, and also if there is evidence of fetal damage by ultrasound
maternal infection but does not exclude the possibility of an acute examination (e.g., hydrocephalus and/or calcifications). In a prospec-
infection acquired early in pregnancy; this is especially true in those tive cohort study conducted in France, amniotic fluid PCR was per-
patients who exhibit a rapid decline in their IgM titers during the acute formed in 261 women who had been identified as having an acute
infection. In these cases, the use of other serologic tests (e.g., IgA, IgE, infection during gestation through routine prenatal screening for
AC/HS, avidity) may be of particular help. toxoplasmosis.387 Overall sensitivity and negative predictive value of
A positive IgM test result requires further assessment with confir- the amniotic fluid PCR for the diagnosis of congenital infection were
matory testing at a reference laboratory (see also “Diagnosis of Specific 92.2% (95% CI, 81% to 98%) and 98.1% (95% CI, 95% to 99.5%),
Clinical Entities”).299,316,318,319 The use of confirmatory testing with a respectively. Specificity and the positive predictive value were 100%.387
combination of serologic tests in a reference laboratory has proved Overall sensitivity has been reported by several groups as varying
helpful in discriminating between recently and more distantly acquired from 65% to 92.2%; specificity and the positive predictive value have
infections, and having an expert interpret the results to the patient’s been reported to be 100% by most groups.346,388 Recently, one group
physician has been shown to reduce unnecessary induced abortions proposed a novel and more realistic interpretation of amniotic fluid
among pregnant women reported to have IgM antibodies.299 Women PCR test results according to the pretest probability of infection and
who are informed that they have a positive IgM test titer and that it gestational age at the time of maternal infection388; their analysis
signifies that their offspring will or might be infected often choose revealed that negative test results are more significant than previously
abortion. Unfortunately, a positive IgM test may not necessarily indi- thought, given that transmission only occurs in a percentage of women
cate infection acquired during gestation (a false-positive result or per- at various stages of gestation and that a negative amniotic fluid PCR
sistence of a IgM-positive result in the chronic stage of the infection), further reduces this percentage. Of note, the vast majority of their
and thus, the abortion may not be indicated. It is for this reason that pregnant women, as is the case for all studies reported from Western
confirmatory testing in a reference laboratory has been recommended Europe, received the benefit of prenatal treatment. Thus, caution
by many experts and by the FDA.318 should be exercised when attempting to apply risks of congenital
A number of tests for avidity of Toxoplasma IgG antibodies infection to infants born to women who were not treated during gesta-
have been introduced to help differentiate between recently acquired tion. The preferable time for amniocentesis is at 18 weeks of gestation
and distant infection.310,311,386 Studies of the kinetics of the avidity of or later if acute infection was acquired after 18 weeks. The sensitivity
IgG in pregnant women who have seroconverted during gestation of the amniotic fluid PCR test appears to be lower when the amnio-
have shown that women with high-avidity test results have been centesis is performed before 18 weeks of gestation279 and should be
infected with T. gondii for at least 3 to 5 months. Recently, it has been avoided before 18 weeks for the purpose of diagnosing congenital
demonstrated that, when used as a confirmatory test, along with a toxoplasmosis.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3145

• Routine serologic screening is recommended during pregnancy, regardless of


epidemiologic history or presence of illness during gestation1
• In addition, serologic testing should be performed during pregnancy in the presence of:

Chapter 280  Toxoplasma gondii


o Flulike or unexplained illness
o Lymphadenopathy
o Fetal ultrasound suggestive of congenital infection

Toxoplasma IgG and IgM


Can be performed at nonreference, hospital-based, or
commercial laboratory

IgG neg IgG pos IgM pos or equivocal. Send serum


IgM neg IgM neg to a reference laboratory for
confirmatory testing.4

No serologic evidence • If <18 weeks’ gestation If seroconversion documented or


of Toxoplasma Infection acquired in the reference laboratory confirms acute
infection. Risk of C.T. distant past and before infection acquired during pregnancy:
only if woman gestation. Risk for C.T. 1. Treatment 5 should be promptly
acquires infection essentially zero unless instituted: (a) spiramycin6 if infection
during pregnancy. patient is acquired before 18 weeks’
Counseling should be immunocompromised. gestation or (b) pyrimethamine7 plus
provided on how to • If >18 weeks’ gestation sulfadiazine plus folinic acid 8 if
avoid primary T. It is difficult to establish acquired at or after 18 weeks
gondii infection (see whether infection occurred 2. If safe and feasible, amniotic fluid
Table 280-4). during or before pregnancy obtained at 18 weeks or onward
should be tested for Toxoplasma PCR
3. Fetal ultrasound should be
obtained for the detection of
abnormalities suggestive of C.T.
Follow-up testing 4. Switch spiramycin to
during gestation to • If <18 weeks’ gestation pyrimethamine plus sulfadiazine plus
detect seroconversion2 No further action required folinic acid if amniotic fluid PCR is
• If >18 weeks’ gestation positive or ultrasound is abnormal
Attempt to obtain earlier 5. Consider testing close household
serum for testing or results contacts for the diagnosis of acute
of previous Toxoplasma Toxoplasma infection or
serologic tests obtained toxoplasmosis in individuals at high
before current pregnancy3 risk
If seroconversion and consult reference
detected (i.e., IgG pos, laboratory.
IgM pos), follow IgM-
pos algorithm

FIGURE 280-8  Diagnostic approach and management algorithm of toxoplasmosis during pregnancy. Most of the initial serologic screening
can be accomplished by nonreference or commercial laboratories. Only positive immunoglobulin M results should be considered for additional testing
and consultation with medical experts at a reference laboratory. C.T., congenital toxoplasmosis; IgG, immunoglobulin G; IgM, immunoglobulin M; neg,
negative test result; pos, positive test result.
1
Up to 50% of women who acquire Toxoplasma infection during gestation do not have a known risk factor for acute infection or an illness suggestive
of toxoplasmosis. Thus, to identify all women at risk, serologic screening should be performed in all pregnant women, along with other routine screening
tests.
2
In a recent study from Lyon, France, monthly screening of seronegative pregnant women was reported to significantly decrease the risk of vertical
transmission and of clinical signs at 3 years of age.476
3
Consider consultation with a physician expert in management of toxoplasmosis during pregnancy (e.g., in the United States, Palo Alto Medical
Foundation–Toxoplasma Serology Laboratory [PAMF-TSL], www.pamf.org/serology/; 650-853-4828; e-mail, toxolab@pamf.org or U.S. [Chicago] National
Collaborative Treatment Trial Study [NCCTS]; 773-834-4152).
4
Consider sending serum sample to a reference laboratory (e.g., PAMF-TSL).
5
Treatment regimens vary by country. The pyrimethamine-sulfadiazine–folinic acid regimen should not be offered to any pregnant women before 12
weeks of gestation because of potential teratogenicity. In some centers in Europe, this regimen is offered at 14 weeks of gestation or later; in the United
States, it is recommended at 18 weeks or later.
6
Spiramycin is not commercially available in the United States. It can be obtained at no cost and after consultation (with PAMF-TSL or the NCCTS through
the U.S. Food and Drug Administration.
7
When using pyrimethamine, folic acid should be discontinued from the prenatal multivitamins. Folic acid can potentially counteract the antiparasitic
effect of the drug.
8
Folic acid should not be erroneously used instead of folinic acid.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3146
Maternal IgG antibodies present in the newborn may reflect either Less serious side effects of pyrimethamine include gastrointestinal dis-
past or recent infection in the mother. For this reason, tests for the tress, rash, headaches, and a bad taste in the mouth.
detection of IgA and IgM antibodies are commonly used for the diag- Unless there are circumstances that preclude the use of more
nosis of infection in the newborn (Fig. 280-9). It is essential that than one drug, there is no role for monotherapy in the treatment of
Part III  Infectious Diseases and Their Etiologic Agents

maternal contamination of blood obtained at birth be excluded; serum toxoplasmosis. A second drug, such as sulfadiazine or clindamycin,
samples obtained from peripheral blood and not from the umbilical should be added. Sulfadiazine acts synergistically with pyrimethamine;
cords are preferred. In a retrospective study of 164 untreated infants most other sulfonamides have inferior activity. The patient must
aged 0 to 180 days, in the United States, T. gondii–specific IgM, IgA, maintain a good urine output to prevent crystalluria and oliguria.
and IgE antibodies were demonstrable in 86.6%, 77.4%, and 40.2% of The most common side effects associated with sulfadiazine are skin
the infants, respectively.284 Testing for IgM and IgA antibodies increased rashes, which may be life-threatening,400 and crystal-induced nephro-
the sensitivity of making the diagnosis of congenital toxoplasmosis to toxicity.401 Worsening encephalopathy, hallucinations, or a new onset
93%, compared with testing for IgM or IgA alone. The sensitivity of of psychiatric symptoms in patients with AIDS may be sulfadiazine
the IgM and IgA has been reported to be somewhat lower in cohorts induced and must be considered in the patient who is nonrespon-
of treated (prenatal treatment) infants: 64% to 70% for IgM, 53% to sive to otherwise appropriate anti-Toxoplasma treatment.402 A drug
65% for IgA, and 66% to 81% for both.388 rash with sulfonamide therapy does not necessarily preclude its use
If IgG antibodies are detected but serologic tests for IgM and IgA because successful desensitization protocols have been reported.403,404
antibodies are negative and T. gondii is not isolated, follow-up sero- Clindamycin appears to act by targeting translation in the apico-
logic testing in suspect cases is indicated to attempt to establish the plast of T. gondii.31 Adverse reactions to clindamycin include rash,
diagnosis. Maternally transferred antibodies usually decline and disap- nausea, vomiting, and diarrhea, which may be associated with Clos-
pear within 6 to 12 months. tridium difficile infection. Myopathy with electromyographic abnor-
Studies using the Western blot technique have shown that maternal malities and elevated serum creatine phosphokinase levels have been
and infant sera may recognize different T. gondii antigens when the described.405
infant is congenitally infected.389,390 Combining Western blot with Although clinical experience with TMP-SMX is more limited, this
conventional serologic analysis (i.e., IgG, IgM, and IgA tests) has drug combination, which targets folate metabolism in a manner similar
been reported to be more sensitive for the diagnosis of congenital to pyrimethamine plus sulfadiazine, has documented activity and can
toxoplasmosis at birth and within the first 3 months of life than either be used in patients requiring parenteral therapy.406 Atovaquone com-
test alone.390-392 Recently, three IgM-bands at 75, 90, and 100 kDa (also bined with pyrimethamine or sulfadiazine, or in unusual circum-
known as the “IgM triplet”) have been reported to increase the sensitiv- stances as a single agent, is another less-well-studied alternative for
ity of the diagnosis of congenital toxoplasmosis to 95.8% when com- patients who can be treated with oral therapy. The role of other drugs,
bined with prenatal and serologic neonatal tests.393 including azithromycin, clarithromycin, atovaquone, and dapsone is
Additional diagnostic methods that have been used successfully to less clear; they should only be used as alternatives to the regimens
diagnose the infection in infants are direct demonstration of the organ- described above and should be used in combination with pyrimeth-
ism by isolation in mice or cell culture (e.g., placental tissue, body amine whenever possible.
fluid) and PCR in body fluids (e.g., CSF, blood, and urine).341,394-397 Spiramycin has been used in pregnant women to attempt to
Evaluation of infants with suspected congenital toxoplasmosis should reduce transmission to the fetus; it has not been shown to be effective
always include ophthalmologic examination, radiologic studies (par- for acute therapy, maintenance therapy, or primary prophylaxis of
ticularly to detect the presence of cerebral calcifications; if feasible, CT TE in AIDS patients. There is no evidence that spiramycin is
is preferred over ultrasound), and examination of CSF. A more detailed teratogenic.
discussion of diagnostic procedures in congenitally infected infants is Although drugs used to treat toxoplasmosis in the setting of differ-
available in a chapter by Remington and colleagues.8 ent clinical entities are basically the same, careful attention should be
In the absence of a systematic screening program for pregnant given to the dosing and dosing regimen. Recommended doses in
women, a secondary prevention program that consists of serologic immunocompromised patients are usually higher than those in immu-
testing of all newborns for IgM antibodies against T. gondii has been nocompetent patients. For instance, the recommended dose of pyri-
implemented in Massachusetts.398,399 Using routine screening of all methamine for patients with TE is 50 to 75 mg/day after a loading dose
newborns, congenital infection was confirmed in approximately 1 in of 200 mg, whereas the dose to treat fetal infection during pregnancy
12,000 infants. More than 90% of these were identified only through is 25 to 50 mg/day after a loading dose of 100 mg/day for 2 days in the
neonatal screening and not through initial clinical examination. mother.
In infants with congenital toxoplasmosis or congenital infection, a
rebound in IgG and IgM antibody titers is frequently observed after
discontinuation of therapy. In our experience, such a serologic rebound Therapy Regimens in Specific  
has not been shown to be clinically significant.8 Clinical Entities
Toxoplasmosis in the Immunocompetent
THERAPY Patient
Currently recommended drugs against T. gondii act primarily against Treatment of immunocompetent adults with the lymphadenopathic
the tachyzoite form and thus do not eradicate the encysted form (brady- form is rarely indicated; this form is usually self-limited. One small
zoite). Pyrimethamine is considered to be the most effective anti- randomized trial demonstrated efficacy of a 1-month course of
Toxoplasma agent and, if feasible, should always be included in drug TMP-SMX in reducing lymphadenopathy (65% response vs. 22% for
regimens used against the parasite. Pyrimethamine is a folic acid antag- placebo).407 If visceral disease is clinically overt or symptoms are severe
onist. The most common side effect is dose-related suppression of the or persistent, treatment may be indicated for 2 to 4 weeks, followed by
bone marrow, which may be decreased by concomitant administration reassessment of the patient’s condition (Table 280-2). Infections
of folinic acid (calcium leucovorin). It is not well established how often acquired by laboratory accident or transfusion of blood products are
a blood count should be obtained; a reasonable strategy would be to potentially more severe, and patients who have been infected in these
check a peripheral blood cell and platelet count twice weekly until ways probably should be treated (see Table 280-2).
hematologic parameters have stabilized in a nontoxic range, and then
every 2 to 4 weeks. Folinic acid should be administered concomitantly Toxoplasmosis in the Immunodeficient Patient
to avoid bone marrow suppression. The parenteral form of folinic acid Because experience with treatment of toxoplasmosis in immunodefi-
is well absorbed orally, and 10 to 20 mg of folinic acid (up to 50 mg/ cient patients has been most extensively studied in patients with AIDS,
day is used in AIDS patients) may be given orally (e.g., with orange this section focuses primarily on this group of patients. However,
juice at the same time as the pyrimethamine). Whereas folinic acid does information on treatment in AIDS patients likely can, in large part and
not inhibit the action of pyrimethamine on tachyzoites, folic acid does in the absence of data, be extrapolated directly to other immunodefi-
and should not be used in patients being treated with pyrimethamine. cient patients (see Table 280-2).

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3147

• Screen all newborns born to mothers suspected or confirmed to have acquired T. gondii
infection during gestation
• Consider neonatal serologic screening in newborns born to mothers who were not screened

Chapter 280  Toxoplasma gondii


during gestation1
• In addition, laboratory testing should be performed at birth in the presence of:
o Visual abnormalities (e.g., strabismus, blindness, chorioretinitis)
o Encephalitis, seizures, hydrocephaly, or microcephaly
o Brain or hepatic calcifications
o Unexplained sepsis
o Hepatosplenomegaly
o Pneumonitis
o Anemia, jaundice, petechiae, thrombocytopenia
o Skin rash, diarrhea, hypothermia

• Toxoplasma IgG, IgM, IgA


o Can be performed at nonreference, hospital-based,
or commercial laboratories.
o However, recommend IgG, IgM-ISAGA, and IgA
at reference laboratory2
• If clinical suspicion is high:
o Toxoplasma PCR in peripheral blood, urine, and
CSF3
o Ophthalmologic evaluation by pediatric retinal
specialist
o Hearing evaluation
o Ultrasound or CT (preferred) scan of the brain
o Lumbar puncture for CSF3 examination4

Initial treatment indicated54 Initial treatment not indicated; Treatment and serologic follow-
• Positive results for lgG plus serologic follow-up indicated up not indicated
positive results for: • Positive results for IgG in the
o IgM in serum sample obtained absence of major clinical signs • Newborn:
after 5 days of life and/or plus negative results for: o Negative IgG and
o IgA in serum sample obtained o IgM and o Negative IgM and
after 10 days of life and/or o IgA and, if performed: o Negative IgA
o PCR in peripheral blood, urine, o PCR in peripheral blood, urine,
or CSF and CSF and
• Positive results for IgG plus • Follow-up of serum IgG every 4
o Major clinical signs65 present to 8 weeks, IgG of maternal • Mother
plus origin typically falls by half every o Negative IgG and
6
o Newborn was born to a mother month7 o Negative IgM
who was infected during • Serologic test results in the
gestation mother can aid in the
interpretation of newborn’s
serologies

FIGURE 280-9  Diagnostic approach and management algorithm of the newborn whose mother has been suspected or confirmed to have
acquired toxoplasmosis during gestation. CSF, cerebrospinal fluid; CT, computed tomography; IgG, IgM, and IgA, immunoglobulins G, M, and A,
respectively; ISAGA, immunosorbent agglutination assay; PCR, polymerase chain reaction.
1
Consider consultation with a physician expert in management of toxoplasmosis during pregnancy (e.g., in the United States, Palo Alto Medical
Foundation–Toxoplasma Serology Laboratory [PAMF-TSL], www.pamf.org/serology/; 650-853-4828; e-mail, toxolab@pamf.org or U.S. [Chicago] National
Collaborative Treatment Trial Study, 773-834-4152).
2
Consider sending serum sample to a reference laboratory (e.g., PAMF-TSL).
3
If lumbar puncture is clinically indicated, deemed safe, and feasible.
4
In an attempt to confirm the diagnosis of congenital toxoplasmosis, CSF should be sent for cell count and differential (congenital toxoplasmosis is one
of the few causes of eosinophilic meningitis), protein (congenital toxoplasmosis is one of the few causes of extreme elevation of CSF protein), glucose,
and T. gondii PCR.
5
The recommended regimen is pyrimethamine plus sulfadiazine plus folinic acid (see text).
6
Major clinical signs are referred here: chorioretinitis, brain calcifications, and hydrocephalus.
7
Maternally transferred IgG antibodies usually decline and disappear within 6 to 12 months of life.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3148

TABLE 280-2  Treatment Regimens for Toxoplasmosis in Immunocompetent and Immunocompromised


Patients*
IMMUNOCOMPROMISED PATIENTS, INCLUDING
Part III  Infectious Diseases and Their Etiologic Agents

THOSE WITH AIDS, TRANSPLANTS, CANCER, OR



IMMUNOCOMPETENT PATIENTS TAKING IMMUNOSUPPRESSOR DRUGS‡
Preferred Regimen
Pyrimethamine (PO) 50 mg q12h for 2 days, followed by 25-50 mg daily 200-mg loading dose, followed by 50 (<60 kg)-75 mg/day (≥60 kg)
plus
Folinic acid§ (PO) 10-20 mg daily (during and 1 wk after therapy with 10 to 20 mg daily (up to 50 mg/day) (during and 1 wk after
pyrimethamine) therapy with pyrimethamine)
plus
Sulfadiazine (PO) 1 g q6h 1000 (<60 kg)-1500 mg (≥60 kg) q6h
Preferred Alternative Regimens
Pyrimethamine–folinic acid Same doses as above Same doses as above
plus
Clindamycin (PO or IV) 300 mg q6h 600 mg q6h (up to 1200 mg q6h)
or
Atovaquone (PO) 1500 mg PO twice daily 1500 mg PO twice daily
Trimethoprim-sulfamethoxazole (PO or IV) 10 mg/kg/day (trimethoprim component) divided in 10 mg/kg/day (trimethoprim component) divided in two to three
two to three doses doses (doses as high as 15-20 mg/kg/day have been used)
Alternative Regimens with Limited Supportive Data‖
Pyrimethamine–folinic acid Same doses as above Same doses as above
plus
Clarithromycin (PO) 500 mg q12h 500 mg q12h
or
Dapsone (PO) 100 mg/day 100 mg/day
or
Azithromycin (PO) 900-1200 mg/day 900-1200 mg/day
Sulfadiazine (PO) Same doses as above Same doses as above
plus
Atovaquone (PO) Same doses as above Same doses as above
*Assistance is available for the diagnosis and management of toxoplasmosis at the Palo Alto Medical Foundation–Toxoplasma Serology Laboratory; Palo Alto, CA;
www.pamf.org/serology/; 650-853-4828; toxolab@pamf.org.

Indicated in patients with (1) ocular disease associated with primary infection or reactivation of latent infection; (2) severe disease including myocarditis, myositis,
hepatitis, pneumonia, brain, or skin lesions and lymphadenopathy; (3) persisting symptoms; and (4) laboratory accidents.

After the successful use of a combination regimen during the acute/primary therapy phase, same agents at half doses are usually used for maintenance or secondary
prophylaxis; for pyrimethamine, use 25 mg (<60 kg) to 50 mg/day (>60 kg); for clindamycin, use 600 mg PO every 8 hours.
§
Folinic acid (leucovorin); should always be given with pyrimethamine to minimize toxicity; folic acid must not be used as a substitute for folinic acid.

These agents have been used in clinical studies with small numbers of patients and have response rates lower than the standard regimens (see text for references). They
should be used only in patients who are intolerant of the standard regimens.
AIDS, acquired immunodeficiency syndrome; IV, intravenous; PO, orally.

If left untreated, toxoplasmosis in immunodeficient patients is often toxoplasmosis and is the standard to which experimental regimens
lethal. Treatment is recommended for 4 to 6 weeks after the resolution should be compared. This regimen is associated with clinical response
of all signs and symptoms (often for 6 months or longer). At one in 68% to 95% of patients with TE.409,410 Unfortunately, up to 40% of
medical center, 80% of non-AIDS immunodeficient patients with toxo- patients develop side effects from one or more of the drugs, often
plasmosis improved with specific therapy.408 This rate of improvement requiring discontinuation of one or both agents.198,356 Pyrimethamine-
is similar to that observed in appropriately treated AIDS patients with clindamycin and folinic acid appear comparable in efficacy to
TE.365,409 Chronic (latent) asymptomatic infection in immunodeficient pyrimethamine-sulfadiazine,409,411 but this combination also has sub-
patients is not treated. stantial toxicity. TMP-SMX412-414 (at 10 mg/kg/day of the TMP compo-
The exact dosing schedule for the treatment of toxoplasmosis in nent, divided in two doses) has shown efficacy similar to the
non-AIDS immunocompromised patients has not been defined. pyrimethamine-sulfadiazine regimen (with a more rapid radiologic
However, useful information in this regard has resulted from studies response in the TMP-SMX group) in a randomized pilot trial in 77
performed in AIDS patients with toxoplasmosis. patients with AIDS406; this provides an alternative regimen for situa-
Therapy for toxoplasmosis in AIDS patients includes acute tions in which parenteral therapy is required or when pyrimethamine
(primary or induction) treatment, maintenance treatment (secondary is unavailable. An international, noncomparative study of atova-
prophylaxis), and primary prophylaxis. There are no convincing data quone415,416 (administered orally as a suspension) combined with either
from prospective, carefully designed trials to allow the recommenda- pyrimethamine or sulfadiazine as treatment for acute disease demon-
tion of monotherapy for induction, maintenance, or primary prophy- strated 6-week response rates of 75% (21 of 28 patients) for atovaquone-
laxis. Because relapse occurs in up to 80% of cases400 after the pyrimethamine and 82% (9 of 11) for atovaquone-sulfadiazine.417
discontinuation of primary therapy, maintenance therapy is recom- Serum levels of atovaquone may predict response but are not com-
mended for all patients until the CD4 count rises above 200 cells/mm3 mercially available.416 Doses of atovaquone lower than currently rec-
for at least 6 months in response to ART (plasma HIV viral load will ommended for treatment (750 mg bid vs. 1500 mg bid) can often
often be below detection limits during this period). achieve therapeutic levels if not co-administered with efavirenz, which
Acute therapy should be for at least 3 weeks,86 and up to 6 weeks can reduce atovaquone levels by approximately 45%.417a TMP-SMX and
or more may be required for more severely ill patients who have not atovaquone both are active against Pneumocystis, and thus anti-
achieved a complete response. Pyrimethamine combined with sulfa- Pneumocystis prophylaxis does not need to be administered when
diazine and folinic acid is the therapy of choice for AIDS patients with these drugs are used. Because clindamycin-pyrimethamine has no

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3149
409
demonstrated anti-Pneumocystis activity, additional prophylaxis (e.g., has been associated with the lowest relapse rate and is recommended
aerosolized pentamidine) needs to be co-administered if patients are unless there are contraindications to its use. When daily therapy with
at risk for developing Pneumocystis pneumonia. pyrimethamine-sulfadiazine was compared with a twice-weekly regi-
In a 13-patient pilot study of TE using the combination of pyri- men for the prevention of recurrence of TE, the latter was found to be

Chapter 280  Toxoplasma gondii


methamine, 75 mg/day, and clarithromycin, 1 g every 12 hours, 62% less effective.426,427 Although a subsequent trial by the same group
of patients had a complete and 23% a partial clinical response; 15% of found that three-times weekly therapy was equivalent to daily therapy,
patients died by week 3 of therapy,418 and adverse events resulted in the relapse rates for both groups (≈14.5/100 patient-years) was higher
discontinuation of therapy in 27%. No long-term follow-up data are than was seen with daily therapy (4.4/100 patient-years) in the earlier
available, and no additional studies with this drug combination have study.427,428 Patients receiving the pyrimethamine-sulfadiazine combi-
been published. Doses of clarithromycin higher than 500 mg twice nation do not require another regimen for PCP prophylaxis. Whereas
daily have been associated with increased mortality in HIV-infected 25% of patients receiving pyrimethamine-clindamycin subsequently
patients receiving therapy for Mycobacterium avium infection and developed PCP,429 no patient receiving pyrimethamine-sulfadiazine
should not be used.419,420 developed PCP.427,429 One 17-patient study demonstrated that TMP-
Dapsone in combination with pyrimethamine has been reported SMX could be safely substituted for pyrimethamine-sulfadiazine after
anecdotally to be effective for the treatment of TE when used in an a median of 24-months maintenance therapy in patients also receiving
oral dose of 100 mg/day with 25 mg/day of oral pyrimethamine.421 ART; the major benefit was in decreasing pill burden.430
Doxycycline has had success in the treatment of TE in a few patients Because of drug toxicity, many patients are unable to continue
when used at 300 mg/day IV in three divided doses.422 A dosage taking the pyrimethamine-sulfadiazine combination for maintenance
of 100 mg twice a day was given to six patients intolerant to therapy. A higher relapse rate has been reported with the use
pyrimethamine-sulfadiazine, but five had associated neurologic and of pyrimethamine-clindamycin compared with pyrimethamine-
radiologic recurrences while receiving the drug.423 Further studies are sulfadiazine for secondary prophylaxis of TE; hence, it is recommended
needed to compare the relative efficacy and toxicity of these alternative that the clindamycin dose be 1800 mg/day if tolerated.409,431,432 Encour-
regimens. Although azithromycin plus pyrimethamine is effective for aging results have been reported with other drug combinations. These
the treatment of some cases of TE in AIDS patients, its use should be include Fansidar (pyrimethamine-sulfadoxine), which has been used
limited, based on results of a recent study demonstrating an inferior in a dose of one tablet twice weekly,433 and pyrimethamine-dapsone
response rate, especially during maintenance therapy.424,425 Alternative administered on an intermittent schedule (two to three times a
regimens used for acute therapy and their dosage schedules are listed week).434-436 The long half-life of these agents allowed the longer dosing
in Table 280-2. interval, but Fansidar may have an increased risk of severe cutaneous
Corticosteroids are often given to patients with TE for the reduction reactions, and the longer half-life results in slower drug clearance after
of cerebral edema and raised intracranial pressure. The clinical response discontinuation of therapy.437 When pyrimethamine was used alone as
and survival in patients with TE who received corticosteroids in addi- maintenance therapy at 50 mg/day438,439 and 100 mg/day,438 the relapse
tion to antimicrobial therapy has been reported to be no different from rates were 10% to 28% and 5%, respectively. Atovaquone alone or in
that of those who received antimicrobial agents alone.365 The use of combination regimens also appears to have activity based on uncon-
these agents may complicate the interpretation of empirical therapy of trolled trials; combination therapy (with sulfadiazine or pyrimeth-
TE because partial clinical and radiologic improvement may be seen amine) should be used whenever possible.415-417,440
solely resulting from a reduction in cerebral edema and inflammation The optimal time to initiate ART in HIV-infected patients with TE
or a response of CNS lymphoma; moreover, they may further compro- has not been well defined. In one randomized trial of 282 patients with
mise the immune systems of these already very immunodeficient opportunistic infections other than tuberculosis (only 5% with TE),
patients. Their use thus should be limited to situations where clinically which compared early (median, 12 days after initiation of opportunis-
significant edema or a mass effect is present. tic infection therapy) to deferred (median, 45 days) initiation of ART,
Seizures occur in up to 35% of patients with TE.356 One retrospec- the secondary end point of AIDS progression or death occurred sig-
tive study demonstrated a poorer outcome in those patients who nificantly less frequently in the early initiation group.441 Thus, in the
received anticonvulsant therapy, compared with those who did not.400 absence of contraindications, ART can reasonably be started 2 to 3
Whether this result represents a true drug effect or a selection bias, weeks after diagnosis of TE.
given that those receiving anticonvulsant therapy are likely to be more Primary prophylaxis against T. gondii in patients with AIDS has
severely ill, is unclear. Anticonvulsant agents may be responsible for been shown to be effective in preventing acute TE.442-445 In addition,
numerous side effects and drug interactions; for instance, potentially the use of new highly active ART in HIV-infected patients has had a
serious interactions can occur between agents such as carbamazepine, profound effect in decreasing the incidence of TE in these patients.87,88
phenobarbital, or phenytoin and other drugs used to treat HIV infec- Primary prophylaxis is recommended for patients who have detectable
tion, such as protease inhibitors. Anticonvulsant therapy is probably Toxoplasma IgG antibodies and whose lowest CD4+ count has been less
best administered only when seizures have occurred.400 than 100/mm3 (many experts use less than 200/mm3 as the cutoff
The time to clinical response in AIDS patients with TE who were rather than 100/mm3), regardless of the HIV RNA viral load.432
receiving appropriate anti-Toxoplasma therapy has been evaluated in a TMP-SMX (1 double-strength or single-strength tablet/day), dapsone
study that included an objective, graded neurologic examination.365 Of (50 mg/day) plus pyrimethamine (50 mg/week), and Fansidar (twice
those with a response, 91% improved with respect to at least half of weekly) have been reported to be effective in preventing the first
their baseline abnormalities by day 14.365 AIDS patients with presumed episode of TE.432,442,445,446
TE had some degree of improvement within 7 to 10 days of the initia- Studies have demonstrated that it is safe to discontinue primary or
tion of appropriate anti-Toxoplasma therapy. By contrast, a significant secondary anti-Toxoplasma prophylaxis when the recovery of a CD4+
number of patients with an alternative diagnosis, including lymphoma, count greater than 200 cells/mm3 is achieved and sustained in patients
exhibited signs of clinical deterioration as early as 3 to 5 days after the on ART.251-253,447,448 Current recommendations are to discontinue pro-
initiation of the empirical regimen for presumed TE.365 Headaches and phylaxis when the CD4 count has risen above 200 cells/mm3 for at least
seizures were insensitive indicators for a response to therapy. In some 3 months (primary prophylaxis) or 6 months (secondary prophy-
cases, toxoplasmosis has progressed to death despite the use of appro- laxis).432 It is important to recognize that in most of these studies, the
priate drug regimens.409,411 median CD4 count was greater than 300 cells/mm3 at enrollment, and
After successful primary therapy, drug dosages are generally viral loads were below detection limits or reasonably controlled in
decreased for maintenance therapy. No single maintenance regimen most patients.
that is efficacious with an acceptable adverse reaction profile has yet At present, TMP-SMX is used by most transplant teams as prophy-
been identified. Relapse of TE occurs in approximately 20% to 30% of laxis against Pneumocystis pneumonia. Its use has also been shown to
patients who are receiving maintenance therapy, in part because of protect against toxoplasmosis. However, TMP-SMX is not protective
nonadherence to and patient intolerance of the prescribed regimen.198 in every case, and some patients are not able to tolerate the drug com-
Pyrimethamine, 25 mg/day, plus sulfadiazine, 500 mg four times daily, bination. In addition, in some patients sulfonamides may be

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3150
contraindicated. Thus, use of TMP-SMX alone may be sufficient for Given the high relapse rate seen in some patients with ocular
prevention of toxoplasmosis in patients who are seronegative for T. toxoplasmosis, prevention of recurrences would be highly desirable.
gondii anti­bodies and who receive heart transplants from seropositive A randomized, open-label trial of 124 patients in Brazil found that
donors (i.e., D+/R− patients).449 The optimal schedule for administra- TMP-SMX (1 double-strength tablet every 3 days) was effective in
Part III  Infectious Diseases and Their Etiologic Agents

tion of TMP-SMX in heart transplant patients has not been defined. decreasing the frequency of recurrences from 24% to 7% in a popu-
Physicians must decide whether a schedule of daily administration or lation at high risk for recurrences.180 If confirmed in other popula-
administration three times each week is to be used. We routinely rec- tions, such a regimen could be beneficial in patients with frequent
ommend daily use of TMP-SMX whenever feasible.10 or severe recurrences. A follow-up report noted that once TMP-SMX
Roxithromycin, administered at 900 mg once a week (may be was stopped, recurrence rates in both groups were similar over a
given in three divided doses),450 has been reported in a small random- 10-year period, suggesting that benefit was seen only while continu-
ized trial to be effective for primary prophylaxis; roxithromycin is ing therapy.467a
unavailable in the United States. Based on a number of clinical trials, For the approach to ocular toxoplasmosis during pregnancy see
pyrimethamine alone cannot be recommended for primary prophy- “Acute Acquired Toxoplasma Infection in Pregnant Women.”
laxis.84,451,452 Clarithromycin453 and spiramycin454 have been ineffective
for primary prophylaxis when they were used alone. In a randomized, Acute Acquired Toxoplasma Infection
placebo-controlled, primary prophylaxis trial, clindamycin (600 mg/ in Pregnant Women
day) was associated with an unacceptably high rate of associated gas- Treatment of the acutely infected pregnant woman does not eliminate
trointestinal disease, in particular diarrhea.455 but does appear to decrease the incidence and severity of fetal infec-
Data on the outcome of treatment of AIDS patients with toxoplas- tion. Because there is usually a delay between the acquisition of acute
mosis outside the CNS are limited; available information on the maternal infection, infection of the placenta, and subsequent infection
therapy of ocular243,456,457 and pulmonary involvement237,458 indicates of the fetus, identification of acute maternal infection necessitates
that these forms of toxoplasmosis are also responsive to treatment. immediate institution of treatment of the mother. Most experience of
Therapy was successful in 50% to 77% of patients with pulmonary maternal treatment to prevent transmission to the fetus has been with
toxoplasmosis.237,458 spiramycin (3 g/day, obtainable in the United States from the FDA,
The hematologic toxicity associated with zidovudine and the high 301-796-1400; after hours, 301-796-8210) (Table 280-3). Spiramycin
doses of pyrimethamine used for the treatment of TE are additive. has been accepted by most investigators as being effective in reducing
Other drugs used in treating HIV-infected patients that cause myelo- the frequency of maternal transmission of T. gondii to the fetus by
suppression include ganciclovir, flucytosine, TMP, pentamidine, che- approximately 60%.277,319 Spiramycin is indicated for patients con-
motherapy agents, and IFN-α. firmed or suspected to have been infected before 18 weeks of gestation.
It appears that its maximal efficacy is best achieved when given within
Ocular Toxoplasmosis 8 weeks of seroconversion.280 Spiramcyin should be continued until
The decision to treat active toxoplasmic chorioretinitis should be made delivery, even if results of the amniotic fluid PCR are negative and
based on a complete ophthalmologic evaluation. A recent report from ultrasound examinations are normal. A retrospective study suggested
the American Academy of Ophthalmology highlighted the limited that the combination of spiramycin and TMP-SMX (after the 14th
data that are available from randomized controlled trials with well- week) is effective in reducing transmission during the second trimester,
defined end points demonstrating the benefits of therapy.459 Treatment compared with historical control subjects.468 If spiramycin cannot be
is most likely indicated in the following settings: any decrease in visual used or is not available, it may be replaced by sulfadiazine (with appro-
acuity, macular or peripapillary lesions, lesions greater than one optic priate precautions at term) or clindamycin alone. However, there are
disk diameter, lesions associated with a moderate-to-severe vitreous no data on the efficacy of sulfonamides, including sulfadiazine, or
inflammatory reaction, the presence of multiple active lesions, the clindamycin when these drugs are used for this purpose.
persistence of active disease for more than 1 month, and any ocular Because spiramycin does not reliably cross the placenta,469 if fetal
lesions associated with recently acquired infection. Because the disease infection is documented or highly suspected or maternal infection is
can be self-limited in immunocompetent patients, many clinicians may confirmed or highly suspected of having been acquired at 18 weeks of
not treat small, peripheral retinal lesions that are not immediately gestation or later, the recommended therapeutic regimen is the com-
vision threatening.9,265,460-462 bination of sulfadiazine (initial dose 75 mg/kg, followed by 50 mg/kg
The reported benefits of medical therapy are related primarily to every 12 hours; maximum, 4 g/day), pyrimethamine (50 mg every 12
the clinical presentation.265,460 Because there is so much variation in the hours for 2 days, followed by 50 mg daily), and folinic acid (10 to
clinical manifestations of the retinal disease, and because the disease 20 mg daily, during and 1 week after completion of pyrimethamine
may be self-limited even without treatment, the response to therapy is therapy) (see Table 280-3). Such treatment might be an alternative to
difficult to interpret. The combination of pyrimethamine (100-mg the termination of pregnancy when abortion is not allowed by law or
loading dose given over 24 hours for 2 days, followed by 25 to 50 mg for women who desire to continue their pregnancy. Pyrimethamine
daily) and sulfadiazine (1 g given four times daily for 4 to 6 weeks), should not be used in the first 18 weeks of pregnancy because of a
depending on the clinical response, which is considered “classic” concern for teratogenicity, although some centers in Europe will use
therapy for ocular toxoplasmosis, is the most common drug combina- it as early as 12 to 14 weeks. In this circumstance, if indicated, sulfa­
tion used (see Table 280-2).460 TMP-SMX showed responses similar to diazine plus clindamycin can be considered, although there are no
pyrimethamine-sulfadiazine in a recent randomized, single-blind trial, data on its efficacy in this situation.8 In addition, pyrimethamine-
although the latter regimen was used at lower-than-standard doses.463 sulfadiazine is also recommended for pregnant women in whom a
Two recent, open, randomized trials found no differences in response recently acquired acute infection is highly suspected or confirmed
rates to intravitreal clindamycin plus dexamethasone compared with during the late second or third trimesters; this is due to the high rates
an oral regimen combining pyrimethamine, sulfadiazine, leucovorin, of vertical transmission observed in those stages of gestation and
and prednisone/prednisolone.464,465 should be recommended even though fetal infection may not yet have
Clindamycin (300 mg orally every 6 hours for a minimum of 3 been confirmed.
weeks) has also been used with favorable clinical results.460 Other drugs A group of European investigators reported between 1999 and 2007
that may have activity but have been inadequately studied include that, in their studies, a significant effect of prenatal treatment on the
atovaquone and pyrimethamine plus azithromycin.466,467 risk of vertical transmission and clinical signs of congenital toxoplas-
Systemic corticosteroids are indicated when lesions involve the mosis was not detected.470-473 These results are not surprising because
macula, optic nerve head, or papillomacular bundle. Photocoagulation the studies included very few untreated women in their analysis, most
has been used both for the treatment of active lesions and for prophy- untreated women were infected during the third trimester, and severe
laxis against the spread of lesions because new lesions appear contigu- cases were excluded.474,475 More recently, several studies from Europe
ous to old lesions.460 In some patients, vitrectomy and lentectomy may have consistently reported evidence for an association between early
be necessary. treatment and reduced risk in the incidence and severity of congenital

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3151

TABLE 280-3  Treatment Regimens for TABLE 280-4  Measures to Prevent Primary
Toxoplasmosis during Pregnancy and for Toxoplasma gondii Infection
Congenital Disease* • Avoid contact with materials potentially contaminated with cat feces,

Chapter 280  Toxoplasma gondii


INDICATION DRUG REGIMEN especially handling of cat litter and gardening. Gloves are advised when these
activities are necessary. Because oocysts require 1 to 2 days to mature,
Pregnant Women dispose of all cat feces daily.
Pregnant women suspected Spiramycin (oral)†: 1 g (3 million units) q8h • Disinfect cat litter box with near boiling water for 5 minutes before handling.
or confirmed of having (for a total of 3 g or 9 million units/day) • Avoid mucous membrane contact when handling raw meat.
acquired infection before Spiramycin should be continued until delivery, • Wash hands thoroughly after contact with raw meat.
18 weeks of gestation. even if fetal infection is not detected (e.g., • Kitchen surfaces and utensils that have come in contact with raw meat
Not recommended during negative amniotic fluid PCR test and should be washed.
pregnancy if the fetus has negative follow-up ultrasound) • Freeze meat to –20° C for at least 48 hours.
documented or suspected • Cook meat to 67° C (153° F) or “well done” (meat should not be “pink” in
infection (see below) the center).
• Avoid ingestion of dried, smoked, or cured meat because they may be
Pregnant women at ≥18 Pyrimethamine: 50 mg q12h for 2 days,
infectious.
weeks of gestation and   followed by 50 mg daily
• Wash fruits and vegetables before consumption.
(1) in whom it is suspected plus
• Refrain from skinning animals.
or confirmed that acute Sulfadiazine: 75 mg/kg (first dose), followed
• Avoid drinking untreated water potentially contaminated with oocysts.
infection was acquired at by 50 mg/kg q12h (max., 4 g/day)
• Avoid drinking unpasteurized goat’s milk.
or after 18 weeks of plus
• Avoid eating raw oysters, clams, and mussels.
gestation or (2) who have Folinic acid†: 10-20 mg daily during and for
a positive amniotic fluid 1 wk after pyrimethamine therapy
PCR test or an abnormal Pyrimethamine is teratogenic and should not
ultrasound suggestive of be used during pregnancy before week 18
congenital toxoplasmosis (in some centers in Europe, it is used as early
as week 14). Sulfadiazine should not be used
alone; consider clindamycin plus sulfadiazine.
Congenital Infection
Detailed information on and recommendations for the postnatal treat-
Infants
ment of congenital toxoplasmosis are reviewed elsewhere,8 but we
Congenital infection; Pyrimethamine: 1 mg/kg q12h for 2 days,
treatment regimen is followed by 1 mg/kg/day for 2 or 6 mo,
favor continuous sulfadiazine (50 mg/kg every 12 hours), pyrimeth-
usually recommended   followed by 1 mg/kg/day every Monday, amine (loading dose, 1 mg/kg every 12 hours for 2 days; then begin-
for 1 yr Wednesday, and Friday ning on day 3, 1 mg/kg per day for 2 or 6 months; then this dose every
plus Monday, Wednesday, and Friday), and folinic acid (10 mg three times
Sulfadiazine: 50 mg/kg q12h
plus
weekly during and for 1 week after pyrimethamine therapy) for a
Folinic acid‡: 10 mg three times weekly minimum of 12 months (see Table 280-3).475,478 Other groups have used
Prednisone (if CSF protein ≥1 g/dL or pyrimethamine-sulfadiazine-folinic acid alternated with spiramycin
severe chorioretinitis): 0.5 mg/kg q12h (100 mg/kg/day).8 Serial follow-up to gauge the response of the infant
(until CSF protein <1 g/dL or resolution of
severe chorioretinitis)
to therapy should include neuroradiology, ophthalmologic examina-
tions, and CSF analysis if indicated.8
Older Children
For guidance on therapy in congenital cases, we recommend that
Active disease; treatment is Pyrimethamine: 1 mg/kg q12h (max., 50 mg) physicians contact Dr. Rima McLeod (773-834-4152) at the University
usually continued for for 2 days, followed by 1 mg/kg/day (max.,
1-2 wk beyond resolution 25 mg) of Chicago, where a major study, the National Collaborative Treat-
of clinical manifestations plus ment Trial, on the appropriate management of these cases is being
Sulfadiazine: 75 mg/kg (first dose), followed performed.478 This study has shown that outcomes are substantially
by 50 mg/kg q12h (max., 4 g/day) better for most, but not all, infants treated from the neonatal period
plus
Folinic acid3: 10-20 mg three times weekly for 12 months with pyrimethamine-sulfadiazine and leucovorin, com-
Prednisone (severe chorioretinitis): pared with historical control subjects receiving no or short-course
1 mg/kg/day in two divided doses; max., therapy.478-481 Improvement in intellectual function, regression of reti-
40 mg/day, rapid taper nal lesions, reduction in anticonvulsant drug requirements, and pre-
*Assistance is available for the diagnosis and management of toxoplasmosis at the vention of auditory sequelae appear to be the major benefits of such
Palo Alto Medical Foundation–Toxoplasma Serology Laboratory; Palo Alto, CA; treatment, which was combined with CSF shunting if required.478
www.pamf.org/serology/; 650-853-4828; toxolab@pamf.org; or U.S. (Chicago)
National Collaborative Treatment Trial Study; 773-834-4152. Signs of active infection resolved within weeks of initiation of treat-

Spiramycin is not teratogenic, and it is available in the United States through ment. In a significant number of treated children, cerebral calcifica-
the investigational new drug process at the U.S. Food and Drug Administration tions diminished in size or resolved.481 More recently, they also
(301-796-1600). Prior consultation with medical consultants is required. reported that new central chorioretinal lesions were uncommon (14%)

Folinic acid (leucovorin) should always be given with pyrimethamine to minimize
toxicity; folic acid must not be used as a substitute for folinic acid. in children with congenital toxoplasmosis who have been treated
CSF, cerebrospinal fluid; max., maximum; PCR, polymerase chain reaction. during their first year of life, compared with historical reports of much
higher rates (≥82%) for untreated children or those treated for only
1 month near birth.292

toxoplasmosis.280,282,283,476 Given that recent studies have consistently PREVENTION


suggested a clinically significant benefit,474 and that a significant benefit General Methods
was never ruled out in previous studies, most authorities continue to Prevention is most important in seronegative pregnant women and
recommend spiramycin or pyrimethamine-sulfadiazine for women immunodeficient patients. It is most readily accomplished through
with suspected or confirmed acute T. gondii infection acquired during education of these patients by their personal physicians (Table 280-
gestation. 4). The goal is to avoid the ingestion of and contact with tissue cysts
Pregnant women with toxoplasmic chorioretinitis as a result of or sporulated oocysts. Tissue cysts in meat are made noninfectious
reactivation of chronic disease do not have a higher risk to transmit by heating the meat to 66° C (meat should be cooked to “well done”
the parasite to their offspring than do pregnant women who have been with no pink meat visible in the center), or by freezing it to −20° C
infected before pregnancy and do not have ocular disease.477 Their eye (which is not attainable in most home freezers) for at least 48 hours.
disease should be treated according to the indications discussed in the Meat that is smoked, cured in brine, or dried may still be infectious.
section “Ocular Toxoplasmosis.” Pregnant women with toxoplasmic Oysters, clams, or mussels should not be eaten raw. Hands should
chorioretinitis thought to be a manifestation of recently acquired infec- be washed thoroughly after handling raw meat or vegetables, eggs
tion should be treated because of both the eye disease and the risk of should not be eaten raw, and unpasteurized milk (particularly milk
transmission of the infection to their fetus. from goats) should be avoided. Vectors such as flies and cockroaches

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3152

TABLE 280-5  Rates of Congenital Transmission in 270 Women and the Sensitivity and Negative Predictive
Value of Amniotic Fluid PCR for Prenatal Diagnosis of Congenital Toxoplasmosis, According to Gestational
Age at Which Maternal Infection Was Acquired
Part III  Infectious Diseases and Their Etiologic Agents

GESTATIONAL AGE AT MATERNAL AMNIOTIC FLUID PCR


INFECTION* (wk) NO. OF INFECTED FETUSES†/TOTAL (%) Sensitivity (CI) (%) NPV (CI) (%)
≤6 0/14 (0) N/A 100 (78-100)
7-11 7/50 (14) 29 (8-65) 90 (81-98)
12-16 7/61 (11.5) 57 (21-94) 95 (86-98)
17-21 14/66 (21.2) 93 (68-99) 98 (90-99.7)
22-26 16/36 (44.4) 63 (39-86) 77 (61-93)
27-31 19/30 (63.3) 68 (48-89) 65 (42-87)
≥32 12/13 (92.3) 50 (22-78) 14 (3-52)
TOTAL 75/270 (28) N/A N/A
*Maternal infection was diagnosed by seroconversion in the 270 women; 261 (97%) were given treatment with spiramycin.

Congenital infection was diagnosed by the persistence of Toxoplasma immunoglobulin antibodies after 1 year of life.
CI, confidence interval; N/A, not applicable; NPV, negative predictive value; PCR, polymerase chain reaction.
Note: The positive predictive value was 100%, regardless of gestational age.
Modified from Montoya JG, Remington JS. Management of Toxoplasma gondii infection during pregnancy. Clin Infect Dis. 2008;47:554-566.

should be controlled. Areas contaminated with cat feces should be results must be interpreted correctly. Nonreference laboratories can
avoided altogether. Disposable gloves should be worn while dispos- effectively accomplish the initial screening testing by simultaneously
ing of cat litter material, working in the garden, or cleaning a child’s performing IgG and IgM antibody tests. Women with positive results
sandbox. Oocysts are killed if the cat litter pan is soaked in nearly in the initial IgG antibody test should have a test for IgM antibody
boiling water for 5 minutes. If the litter pan is cleaned every day, performed on the same serum. Only IgM-positive test results need to
oocysts will not have a chance to sporulate. Serologic testing of cats be sent to a reference laboratory for confirmatory testing (e.g., PAMF-
is unwarranted because testing does not demonstrate whether the TSL).316,318 Clinical decisions should not be made based on positive
infected cat is excreting oocysts. Untreated water has been shown IgM test results alone. In patients with IgG antibodies at any titer, a
to be an effective vehicle for the transmission of the parasite, and negative IgM antibody test result in the first trimester, and no clinical
drinking water sources potentially contaminated with oocyts should signs of acute toxoplasmosis, no further testing would be necessary
be avoided. because the probability of acute acquired infection in these women is
extremely low. Given the same circumstances in the second trimester of
Serologic Screening and Prophylaxis pregnancy, a negative IgM test result rules out, for practical purposes,
Acute Toxoplasma gondii Infection and recent acquisition of acute infection. A negative IgM test in the third
Toxoplasmosis in the Immunodeficient trimester may occur in a patient who acquired the infection earlier
Patient in gestation.
Transmission of T. gondii and death caused by the infection have In some countries (e.g., France, Austria, Italy, Slovenia, Lithuania,
resulted from the transfusion of leukocyte-rich blood products and by and Uruguay), initially seronegative pregnant women are retested at
organ transplantation in immunodeficient patients. Transmission of various intervals during gestation to detect seroconversion and insti-
infection by these routes may occur frequently enough to warrant tute treatment. Monthly screening of seronegative pregnant women
screening for antibody to T. gondii in leukocyte-rich blood product was recently reported to significantly decrease the risk of vertical
donors and possibly to exclude seropositive people as organ donors to transmission and of clinical signs at 3 years of age.476 The appropri-
seronegative potential recipients whenever feasible. ate use of prenatal diagnosis, followed by the optimal use of spira-
Primary prophylaxis can prevent toxoplasmosis in patients dually mycin and/or pyrimethamine-sulfadiazine-folinic acid can markedly
infected with HIV and T. gondii (see “Toxoplasmosis in the Immuno- reduce the incidence of clinically significant congenital toxoplasmo-
deficient Patient” under “Therapy”). Prophylactic treatment (pyri- sis.* Its absence is often associated with poor outcomes.284 Results of
methamine, 25 mg orally every day for 6 weeks after transplantation) a study in France279 on the incidence of T. gondii infection in fetuses
has been used with apparent success in seronegative recipients of of women whose date of acquiring the infection during the gesta-
hearts transplanted from seropositive donors.482 However, TMP-SMX tion was known and who were treated with spiramycin are shown in
used for PCP prophylaxis in solid-organ transplant patients is also Table 280-5.
effective as primary prophylaxis against T. gondii and can be used In pregnant women infected with both HIV and T. gondii, we rec-
without addition of pyrimethamine. Primary prophylaxis in BMT ommend that primary prophylaxis against T. gondii be introduced
and HSCT patients is particularly challenging because TMP-SMX when their CD4 T-cell count falls below 200/mm3. If the patient is
cannot safely be used early (i.e., before engraftment), whereas in receiving TMP-SMX, additional prophylaxis is probably not necessary.
patients with all other transplant organs, it can be used immediately Otherwise, spiramycin can be used at a dose of 3 g/day.
after transplantation.
ACKNOWLEDGMENTS
Congenital Toxoplasma gondii Infection We gratefully acknowledge and thank Dr. Jack S. Remington, whose
and Toxoplasmosis lifelong study of toxoplasmosis and authorship of previous editions of
Congenital toxoplasmosis is a preventable disease. It is therefore the this chapter provided the foundation upon which the current chapter
responsibility of physicians who care for pregnant women to educate is based. We would also like to thank Dr. Alan Sher for his review of
them on how they can prevent themselves from becoming infected (and the manuscript and helpful suggestions.
thereby not place their fetus at risk). A lack of adoption of a systematic
serologic screening program in the United States leaves education as the
principal means of preventing this tragic disease. If physicians choose
to screen their patients serologically, the appropriate tests must be used,
the laboratory performing the tests must be competent, and the test *References 279, 282, 283, 476, 483, 484.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3153
Key References survey from the Infectious Diseases Working Party of the
European Group for Blood and Marrow Transplantation.
414. Torre D, Speranza F, Martegani R, et al. A retrospective
study of treatment of cerebral toxoplasmosis in AIDS
Bone Marrow Transplant. 2000;25:1111-1114. patients with trimethoprim-sulphamethoxazole. J Infect.
The complete reference list is available online at Expert Consult.
246. Antinori A, Larussa D, Cingolani A, et al. Prevalence, asso- 1998;37:15-18.

Chapter 280  Toxoplasma gondii


ciated factors, and prognostic determinants of AIDS- 416. Torres R, Weinberg W, Stansell J, et al. Atovaquone for
5. Vietzke WM, Gelderman AH, Grimley PM, Valsamis
related toxoplasmic encephalitis in the era of advanced salvage treatment and suppression of toxoplasmic encepha-
MP. Toxoplasmosis complicating malignancy. Experience
highly active antiretroviral therapy. Clin Infect Dis. 2004; litis in patients with AIDS. Clin Infect Dis. 1997;24:422-
at the National Cancer Institute. Cancer. 1968;21:816-
39:1681-1691. 429.
827.
251. Furrer H, Opravil M, Bernasconi E, et al. Stopping primary 417. Chirgwin K, Hafner R, Leport C, et al. Randomized phase
7. Luft BJ, Conley F, Remington JS, et al. Outbreak of central-
prophylaxis in HIV-1-infected patients at high risk of Toxo- II trial of atovaquone with pyrimethamine or sulfadiazine
nervous-system toxoplasmosis in western Europe and
plasma encephalitis. Swiss HIV Cohort Study. Lancet. 2000; for treatment of toxoplasmic encephalitis in patients with
North America. Lancet. 1983;1:781-784.
355:2217-2218. acquired immunodeficiency syndrome: ACTG 237/ANRS
8. Remington JS, McLeod R, Thulliez P, Desmonts G. Toxo-
252. Kirk O, Reiss P, Uberti-Foppa C, et al. Safe interruption of 039 Study. AIDS Clinical Trials Group 237/Agence Natio-
plasmosis. In: Remington JS, Klein JO, Wilson CB, et al,
maintenance therapy against previous infection with four nale de Recherche sur le SIDA, Essai 039. Clin Infect Dis.
eds. Infectious Diseases of the Fetus and Newborn Infant. 7th
common HIV-associated opportunistic pathogens during 2002;34:1243-1250.
ed. Philadelphia: Saunders; 2011:918-1041.
potent antiretroviral therapy. Ann Intern Med. 2002;137: 417a.  Calderón MM, Penzak SR, Pau AK, et al. Efavirenz but
19. Su C, Khan A, Zhou P, et al. Globally diverse Toxoplasma
239-250. not atazanavir/ritonavir significantly reduces atovaquone
gondii isolates comprise six major clades originating from
264a.  Jones JL, Bonetti V, Holllan GN, et al. Ocular toxoplasmo- concentrations in HIV-infected subjects. Clin Infect Dis.
a small number of distinct ancestral lineages. Proc Natl
sis in the United States: recent and remote infections. Clin 2016;62:1036-1042.
Acad Sci U S A. 2012;109:5844-5849.
Infect Dis. 2015;60:271-273. 427. Podzamczer D, Miró J, Bolao F, et al. Twice-weekly main-
49. Hill D, Coss C, Dubey JP, et al. Identification of
267. Kong JT, Grigg ME, Uyetake L, et al. Serotyping of Toxo- tenance therapy with sulfadiazine-pyrimethamine to
a sporozoite-specific antigen from Toxoplasma gondii.
plasma gondii infections in humans using synthetic pep- prevent recurrent toxoplasmic encephalitis in patients with
J Parasitol. 2011;97:328-337.
tides. J Infect Dis. 2003;187:1484-1495. AIDS. Ann Intern Med. 1995;123:175-180.
65. Bowie WR, King AS, Werker DH, et al. Outbreak of
278. Hohlfeld P, Daffos F, Thulliez P, et al. Fetal toxoplasmosis: 429. Heald A, Flepp M, Chave J-P, et al. Treatment for cerebral
toxoplasmosis associated with municipal drinking water.
outcome of pregnancy and infant follow-up after in utero toxoplasmosis protects against Pneumocystis carinii pneu-
Lancet. 1997;350:173-177.
treatment. J Pediatr. 1989;115:765-769. monia in patients with AIDS. Ann Intern Med. 1991;115:
69. Bahia-Oliveira LM, Jones JL, Azevedo-Silva J, et al.
281. Dunn D, Wallon M, Peyron F, et al. Mother-to-child trans- 760-763.
Highly endemic, waterborne toxoplasmosis in north
mission of toxoplasmosis: risk estimates for clinical coun- 432. Kaplan JE, Benson C, Holmes KH, et al. Guidelines for
Rio de Janeiro state, Brazil. Emerg Infect Dis. 2003;9:
selling. Lancet. 1999;353:1829-1833. prevention and treatment of opportunistic infections in
55-62.
283. Cortina-Borja M, Tan HK, Wallon M, et al. Prenatal treat- HIV-infected adults and adolescents: recommendations
79. Jones JL, Kruszon-Moran D, Sanders-Lewis K, Wilson M.
ment for serious neurological sequelae of congenital toxo- from CDC, the National Institutes of Health, and the HIV
Toxoplasma gondii infection in the United States, 1999
plasmosis: an observational prospective cohort study. PLoS Medicine Association of the Infectious Diseases Society
2004, decline from the prior decade. Am J Trop Med Hyg.
Med. 2010;7. pii:e1000351. of America. MMWR Recomm Rep. 2009;58:1-207; quiz
2007;77:405-410.
291. Phan L, Kasza K, Jalbrzikowski J, et al. Longitudinal study CE1-4.
83. Slavin MA, Meyers JD, Remington JS, Hackman RC. Toxo-
of new eye lesions in children with toxoplasmosis who were 440. Katlama C, Mouthon B, Gourdon D, et al. Atovaquone as
plasma gondii infection in marrow transplant recipients:
not treated during the first year of life. Am J Ophthalmol. long-term suppressive therapy for toxoplasmic encephalitis
a 20 year experience. Bone Marrow Transplant. 1994;13:
2008;146:375-384. in patients with AIDS and multiple drug intolerance. AIDS.
549-557.
292. Phan L, Kasza K, Jalbrzikowski J, et al. Longitudinal study 1996;10:1107-1112.
87. Abgrall S, Rabaud C, Costagliola D. Incidence and risk
of new eye lesions in treated congenital toxoplasmosis. 441. Zolopa A, Andersen J, Powderly W, et al. Early antiretrovi-
factors for toxoplasmic encephalitis in human immuno­
Ophthalmology. 2008;115:553-559.e8. ral therapy reduces AIDS progression/death in individuals
deficiency virus-infected patients before and during the
300. Montoya JG, Liesenfeld O, Kinney S, et al. VIDAS test for with acute opportunistic infections: a multicenter random-
highly active antiretroviral therapy era. Clin Infect Dis.
avidity of Toxoplasma-specific immunoglobulin G for con- ized strategy trial. PloS One. 2009;4:e5575.
2001;33:1747-1755.
firmatory testing of pregnant women. J Clin Microbiol. 442. Carr A, Tindall B, Brew BJ, et al. Low-dose trimethoprim-
128. Suzuki Y, Orellana MA, Schreiber RD, Remington JS.
2002;40:2504-2508. sulfamethoxazole prophylaxis for toxoplasmic encephalitis
Interferon-γ: the major mediator of resistance against Toxo-
319. Montoya JG, Remington JS. Management of Toxoplasma in patients with AIDS. Ann Intern Med. 1992;117:106-111.
plasma gondii. Science. 1988;240:516-518.
gondii infection during pregnancy. Clin Infect Dis. 2008;47: 445. Girard P-M, Landman R, Gaudebout C, et al. Dapsone-
179. Montoya JG, Parmley S, Liesenfeld O, et al. Use of the
554-566. pyrimethamine compared with aerosolized pentamidine as
polymerase chain reaction for diagnosis of ocular toxoplas-
350a.  Robert-Gangneux F, Sterkers Y, Yera H, et al. Molecular a primary prophylaxis against Pneumocystis carinii pneu-
mosis. Ophthalmology. 1999;106:1554-1563.
diagnosis of toxoplasmosis in immunocompromised monia and toxoplasmosis in HIV infection. N Engl J Med.
180. Silveira C, Belfort R Jr, Muccioli C, et al. The effect of long-
patients: a three-year multicenter retrospective study. J Clin 1993;328:1514-1520.
term intermittent trimethoprim/sulfamethoxazole treat-
Microbiol. 2015;53:1677-1684. 460. Holland GN, Lewis KG. An update on current practices in
ment on recurrences of toxoplasmic retinochoroiditis. Am
356. Porter SB, Sande M. Toxoplasmosis of the central nervous the management of ocular toxoplasmosis. Am J Ophthal-
J Ophthalmol. 2002;134:41-46.
system in the acquired immunodeficiency syndrome. N mol. 2002;134:102-114.
181. Jones JL, Muccioli C, Belfort R Jr, et al. Recently acquired
Engl J Med. 1992;327:1643-1648. 463. Soheilian M, Sadoughi MM, Ghajarnia M, et al. Prospec-
Toxoplasma gondii infection, Brazil. Emerg Infect Dis.
357. Haverkos HW. Assessment of therapy for toxoplasma enceph- tive randomized trial of trimethoprim/sulfamethoxazole
2006;12:582-587.
alitis. The TE Study Group. Am J Med. 1987;82:907-914. versus pyrimethamine and sulfadiazine in the treatment
182. Gilbert RE, Freeman K, Lago EG, et al. Ocular sequelae of
358. Levy RM, Rosenbloom S, Perrett LV. Neuroradiologic find- of ocular toxoplasmosis. Ophthalmology. 2005;112:1876-
congenital toxoplasmosis in Brazil compared with Europe.
ings in AIDS: a review of 200 cases. Am J Neuroradiol. 1882.
PLoS Negl Trop Dis. 2008;2:e277.
1986;147:977-983. 467a.  Silveira C, Muccioli C, Nussenblatt R, et al. The effect of
202. McCabe RE, Brooks RG, Dorfman RF, Remington JS.
365. Luft BJ, Hafner R, Korzun AH, et al. Toxoplasmic encepha- long-term intermittent trimethoprim/sulfamethoxazole
Clinical spectrum in 107 cases of toxoplasmic lymphade-
litis in patients with the acquired immunodeficiency syn- treatment on recurrences of toxoplasmic retinochoroiditis:
nopathy. Rev Infect Dis. 1987;9:754-774.
drome. N Engl J Med. 1993;329:995-1000. 10 years of follow-up. Ocul Immunol Inflamm.
208. Burnett AJ, Shortt SG, Isaac-Renton J, et al. Multiple cases
372. Navia BA, Petito CK, Gold JW, et al. Cerebral toxoplasmo- 2015;23:246-247.
of acquired toxoplasmosis retinitis presenting in an out-
sis complicating the acquired immune deficiency syn- 476. Wallon M, Peyron F, Cornu C, et al. Congenital Toxo-
break. Ophthalmology. 1998;105:1032-1037.
drome: clinical and neuropathological findings in 27 plasma infection: monthly prenatal screening decreases
223. Montoya JG, Giraldo LF, Efron B, et al. Infectious compli-
patients. Ann Neurol. 1986;19:224-238. transmission rate and improves clinical outcome at age 3
cations among 620 consecutive heart transplant patients
409. Katlama C, De Wit S, O’Doherty E, et al. Pyrimethamine- years. Clin Infect Dis. 2013;56:1223-1231.
at Stanford University Medical Center. Clin Infect Dis.
clindamycin vs. pyrimethamine-sulfadiazine as acute and 483. Daffos F, Forestier F, Capella-Pavlovsky M, et al. Prenatal
2001;33:629-640.
long-term therapy for toxoplasmic encephalitis in patients management of 746 pregnancies at risk for congenital toxo-
231. Martino R, Bretagne S, Rovira M, et al. Toxoplasmosis after
with AIDS. Clin Infect Dis. 1996;22:268-275. plasmosis. N Engl J Med. 1988;318:271-275.
hematopoietic stem transplantation. Report of a 5-year

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3153.e1
References peroxidase-antiperoxidase method to demonstrate Toxo-
plasma in formalin fixed, paraffin embedded tissue sec-
from a farm in Massachusetts. J Parasitol. 2002;88:1234-
1238.
1. Montoya JG, Liesenfeld O. Toxoplasmosis. Lancet. 2004;
tions. Hum Pathol. 1981;12:690-698. 60. Dubey JP, Sundar N, Hill D, et al. High prevalence and
363:1965-1976.
30. McFadden GI. The apicoplast. Protoplasma. 2011;248:641- abundant atypical genotypes of Toxoplasma gondii isolated
2. Nicolle C, Manceaux L. Sur une infection a corps de Leish-

Chapter 280  Toxoplasma gondii


650. from lambs destined for human consumption in the USA.
man (ou organismes voisins) du gondi. C R Seances Acad
31. Camps M, Arrizabalaga G, Boothroyd J. An rRNA muta- Int J Parasitol. 2008;38:999-1006.
Sci. 1908;146:207-209.
tion identifies the apicoplast as the target for clindamycin 61. Riemann HP, Meyer ME, Theis JH, et al. Toxoplasmosis in
3. Janku J. Pathogenesa a pathologicka anatomie tak naz-
in Toxoplasma gondii. Mol Microbiol. 2002;43:1309-1318. an infant fed unpasteurized goat milk. J Pediatr. 1975;87:
vaneho vrozeneho kolobomu zlute skvrny v oku normalne
32. Yeh E, DeRisi JL. Chemical rescue of malaria parasites 573-576.
velikem a mikrophthalmickem s nalezem parazitu v sitnici.
lacking an apicoplast defines organelle function in blood- 62. Sacks JJ, Roberto RR, Brooks NF. Toxoplasmosis infection
Cas Lek Ces. 1923;62:1021-1027, 54-59, 81-85, 111-115,
stage Plasmodium falciparum. PLoS Biol. 2011;9:e1001138. associated with raw goat’s milk. JAMA. 1982;248:1728-
138-144.
33. Remington JS, Cavanaugh EN. Isolation of the encysted 1732.
4. Wolf A, Cowen D, Paige BH. Toxoplasmic encephalomy-
form of Toxoplasma gondii from human skeletal muscle 63. Dubey JP, Graham DH, Blackston CR, et al. Biological and
elitis, III. A new case of granulomatous encephalomyelitis
and brain. N Engl J Med. 1965;273:1308-1310. genetic characterisation of Toxoplasma gondii isolates from
due to a protozoon. Am J Pathol. 1939;15:657-694.
34. Dubey J, Thayer D. Killing of different strains of Toxo- chickens (Gallus domesticus) from São Paulo, Brazil: unex-
5. Vietzke WM, Gelderman AH, Grimley PM, Valsamis MP.
plasma gondii tissue cysts by irradiation under defined pected findings. Int J Parasitol. 2002;32:99-105.
Toxoplasmosis complicating malignancy. Experience at the
conditions. J Parasitol. 1994;80:764-767. 64. Swartzberg JE, Remington JS. Transmission of Toxoplasma.
National Cancer Institute. Cancer. 1968;21:816-827.
35. Dubey J, Kotula A, Sharar A, et al. Effect of high tempera- Am J Dis Child. 1975;129:777-779.
6. Israelski DM, Remington JS. Toxoplasmosis in the non-
ture on infectivity of Toxoplasma gondii tissue cysts in pork. 65. Bowie WR, King AS, Werker DH, et al. Outbreak of
AIDS immunocompromised host. In: Remington J, Swartz
J Parasitol. 1990;76:201. toxoplasmosis associated with municipal drinking water.
M, eds. Current Clinical Topics in Infectious Diseases. vol.
36. Jacobs L, Remington JS, Melton ML. The resistance of the Lancet. 1997;350:173-177.
13. London: Blackwell Scientific; 1993:322-356.
encysted form of Toxoplasma gondii. J Parasitol. 1960;46: 66. Luft BJ, Remington JS. Acute Toxoplasma infection among
7. Luft BJ, Conley F, Remington JS, et al. Outbreak of central-
11-21. family members of patients with acute lymphadenopathic
nervous-system toxoplasmosis in western Europe and
37. El-Nawawi FA, Tawfik MA, Shaapan RM. Methods for toxoplasmosis. Arch Intern Med. 1984;144:53-56.
North America. Lancet. 1983;1:781-784.
inactivation of Toxoplasma gondii cysts in meat and tissues 67. Masur H, Jones TC, Lempert JA, Cherubini TD. Outbreak
8. Remington JS, McLeod R, Thulliez P, Desmonts G. Toxo-
of experimentally infected sheep. Foodborne Pathog Dis. of toxoplasmosis in a family and documentation of
plasmosis. In: Remington JS, Klein JO, Wilson CB, et al,
2008;5:687-690. acquired retinochoroiditis. Am J Med. 1978;64:396-402.
eds. Infectious Diseases of the Fetus and Newborn Infant. 7th
38. Ferguson DJP, Graham DI, Hutchinson WM. Pathological 68. Miller MA, Gardner IA, Kreuder C, et al. Coastal freshwa-
ed. Philadelphia: Saunders; 2011:918-1041.
changes in the brains of mice infected with Toxoplasma ter runoff is a risk factor for Toxoplasma gondii infection
9. Wakefield D, Cunningham ET Jr, Pavesio C, et al. Contro-
gondii: a histological immunocytochemical and ultra struc- of southern sea otters (Enhydra lutris nereis). Int J Parasitol.
versies in ocular toxoplasmosis. Ocul Immunol Inflamm.
tural study. Int J Exp Path. 1991;72:463-474. 2002;32:997-1006.
2011;19:2-9.
39. Sims TA, Hay J, Talbot IC. An electron microscope and 69. Bahia-Oliveira LM, Jones JL, Azevedo-Silva J, et al. Highly
10. Derouin F, Pelloux H. Prevention of toxoplasmosis in trans-
immunohistochemical study of the intracellular location endemic, waterborne toxoplasmosis in north Rio de
plant patients. Clin Microbiol Infect. 2008;14:1089-1101.
of Toxoplama tissue cysts within the brains of mice with Janeiro state, Brazil. Emerg Infect Dis. 2003;9:55-62.
11. Dubey JP, Lindsay DS, Speer CA. Structures of Toxoplasma
congenital toxoplasmosis. Br J Exp Pathol. 1989;70:317- 70. Baldursson S, Karanis P. Waterborne transmission of pro-
gondii tachyzoites, bradyzoites, and sporozoites and
325. tozoan parasites: review of worldwide outbreaks—an
biology and development of tissue cysts. Clin Microbiol
40. Jones TC, Bienz KA, Erb P. In vitro cultivation of Toxo- update 2004-2010. Water Res. 2011;45:6603-6614.
Rev. 1998;11:267-299.
plasma gondii cysts in astrocytes in the presence of gamma 71. Jones JL, Dubey JP. Waterborne toxoplasmosis—recent
12. Sibley LD, Boothroyd JC. Virulent strains of Toxoplasma
interferon. Infect Immun. 1986;51:146-156. developments. Exp Parasitol. 2010;124:10-25.
gondii comprise a single clonal lineage. Nature. 1992;359:
41. Ferguson DJ, Hutchison WM, Pettersen E. Tissue cyst 72. Dumetre A, Le Bras C, Baffet M, et al. Effects of ozone
82-85.
rupture in mice chronically infected with Toxoplasma and ultraviolet radiation treatments on the infectivity of
13. Howe DK, Honore S, Derouin F, Sibley LD. Determination
gondii. Parasitol Res. 1989;75:599-603. Toxoplasma gondii oocysts. Vet Parasitol. 2008;153:209-
of genotypes of Toxoplasma gondii strains isolated from
42. Koshy AA, Dietrich HK, Christian DA, et al. Toxoplasma 213.
patients with toxoplasmosis. J Clin Microbiol. 1997;35:
co-opts host cells it does not invade. PLoS Pathog. 2012;8: 73. Lindsay DS, Collins MV, Mitchell SM, et al. Sporulation
1411-1414.
e1002825. and survival of Toxoplasma gondii oocysts in seawater.
14. Fuentes I, Rubio JM, Ramirez C, Alvar J. Genotypic char-
43. Dubey JP, Jones JL. Toxoplasma gondii infection in humans J Eukaryot Microbiol. 2003;50(suppl):687-688.
acterization of Toxoplasma gondii strains associated with
and animals in the United States. Int J Parasitol. 2008;38: 74. Wainwright KE, Miller MA, Barr BC, et al. Chemical inac-
human toxoplasmosis in Spain: direct analysis from clinical
1257-1278. tivation of Toxoplasma gondii oocysts in water. J Parasitol.
samples. J Clin Microbiol. 2001;39:1566-1570.
44. Neu HC. Toxoplasmosis transmitted at autopsy. JAMA. 2007;93:925-931.
15. Ajzenberg D, Cogne N, Paris L, et al. Genotype of 86
1967;202:284-285. 75. Vaudaux JD, Muccioli C, James ER, et al. Identification of
Toxoplasma gondii isolates associated with human congeni-
45. Dubey J. Toxoplasmosis. Am Vet Med Assoc. 1994;205: an atypical strain of Toxoplasma gondii as the cause of a
tal toxoplasmosis, and correlation with clinical findings.
1593-1598. waterborne outbreak of toxoplasmosis in Santa Isabel do
J Infect Dis. 2002;186:684-689.
46. Bevins SN, Carver S, Boydston EE, et al. Three pathogens Ivai, Brazil. J Infect Dis. 2010;202:1226-1233.
16. Boothroyd JC, Grigg ME. Population biology of Toxo-
in sympatric populations of pumas, bobcats, and domestic 76. Conrad PA, Miller MA, Kreuder C, et al. Transmission of
plasma gondii and its relevance to human infection: do
cats: implications for infectious disease transmission. PloS Toxoplasma: clues from the study of sea otters as sentinels
different strains cause different disease? Curr Opin Micro-
One. 2012;7:e31403. of Toxoplasma gondii flow into the marine environment. Int
biol. 2002;5:438-442.
47. Desmonts G, Couvreur J, Alison F, et al. [Epidemiological J Parasitol. 2005;35:1155-1168.
17. Lehmann T, Marcet PL, Graham DH, et al. Globalization
study on toxoplasmosis: the influence of cooking slaughter- 77. Smith KL, Wilson M, Hightower AW, et al. Prevalence of
and the population structure of Toxoplasma gondii. Proc
animal meat on the incidence of human infection]. Rev Fr Toxoplasma gondii antibodies in U.S. military recruits in
Natl Acad Sci U S A. 2006;103:11423-11428.
Etud Clin Biol. 1965;10:952-958 [in French]. 1989: comparison with data published in 1965. Clin Infect
18. Darde ML. Toxoplasma gondii, “new” genotypes and viru-
48. Dattoli VC, Veiga RV, Cunha SS, et al. Oocyst ingestion as Dis. 1996;23:1182-1183.
lence. Parasite. 2008;15:366-371.
an important transmission route of Toxoplasma gondii in 78. Rosso F, Les JT, Agudelo A, et al. Prevalence of infection
19. Su C, Khan A, Zhou P, et al. Globally diverse Toxoplasma
Brazilian urban children. J Parasitol. 2011;97:1080-1084. with Toxoplasma gondii among pregnant women in Cali,
gondii isolates comprise six major clades originating from
49. Hill D, Coss C, Dubey JP, et al. Identification of Colombia, South America. Am J Trop Med Hyg. 2008;78:
a small number of distinct ancestral lineages. Proc Natl
a sporozoite-specific antigen from Toxoplasma gondii. 504-508.
Acad Sci U S A. 2012;109:5844-5849.
J Parasitol. 2011;97:328-337. 79. Jones JL, Kruszon-Moran D, Sanders-Lewis K, Wilson M.
20. Minot S, Melo MB, Li F, et al. Admixture and recombina-
50. Jones JL, Dubey JP. Foodborne toxoplasmosis. Clin Infect Toxoplasma gondii infection in the United States, 1999-
tion among Toxoplasma gondii lineages explain global
Dis. 2012;55:845-851. 2004, decline from the prior decade. Am J Trop Med Hyg.
genome diversity. Proc Natl Acad Sci U S A. 2012;109:13458-
51. Hill DE, Dubey JP. Toxoplasma gondii prevalence in farm 2007;77:405-410.
13463.
animals in the United States. Int J Parasitol. 2013;43: 80. Siegel SE, Lunde MN, Gelderman AH, et al. Transmission
21. Fritz HM, Buchholz KR, Chen X, et al. Transcriptomic
107-113. of toxoplasmosis by leukocyte transfusion. Blood. 1971;37:
analysis of Toxoplasma development reveals many novel
52. Elmore SA, Jones JL, Conrad PA, et al. Toxoplasma gondii: 388-394.
functions and structures specific to sporozoites and
epidemiology, feline clinical aspects, and prevention. 81. Ryning FW, McLeod R, Maddox JC, et al. Probable trans-
oocysts. PloS One. 2012;7:e29998.
Trends Parasitol. 2010;26:190-196. mission of Toxoplasma gondii by organ transplantation.
22. Fritz HM, Bowyer PW, Bogyo M, et al. Proteomic analysis
53. Hill D, Dubey JP. Toxoplasma gondii: transmission, diagno- Ann Intern Med. 1979;90:47-49.
of fractionated Toxoplasma oocysts reveals clues to their
sis and prevention. Clin Microbiol Infect. 2002;8:634-640. 82. Mele A, Paterson PJ, Prentice HG, et al. Toxoplasmosis in
environmental resistance. PloS One. 2012;7:e29955.
54. Remington JS. Toxoplasmosis and congenital infection. bone marrow transplantation: a report of two cases and
23. Radke JR, Striepen B, Guerini MN, et al. Defining the cell
Intrauterine Infect. 1968;4:47-56. systematic review of the literature. Bone Marrow Trans-
cycle for the tachyzoite stage of Toxoplasma gondii. Mol
55. Dubey JP. Isolation of Toxoplasma gondii from a naturally plant. 2002;29:691-698.
Biochem Parasitol. 2001;115:165-175.
infected beef cow. J Parasitol. 1992;78:151-153. 83. Slavin MA, Meyers JD, Remington JS, Hackman RC. Toxo-
24. Dubey JP. Advances in the life cycle of Toxoplasma gondii.
56. Aspinall TV, Marlee D, Hyde JE, Sims PF. Prevalence of plasma gondii infection in marrow transplant recipients:
Int J Parasitol. 1998;28:1019-1024.
Toxoplasma gondii in commercial meat products as moni- a 20 year experience. Bone Marrow Transplant. 1994;13:
25. Barragan A, Sibley LD. Transepithelial migration of Toxo-
tored by polymerase chain reaction—food for thought? Int 549-557.
plasma gondii is linked to parasite motility and virulence.
J Parasitol. 2002;32:1193-1199. 84. Leport C, Chêne G, Morlat P, et al. Pyrimethamine for
J Exp Med. 2002;195:1625-1633.
57. Hill DE, Haley C, Wagner B, et al. Seroprevalence of and primary prophylaxis of toxoplasmic encephalitis in patients
26. Lachenmaier SM, Deli MA, Meissner M, Liesenfeld O.
risk factors for Toxoplasma gondii in the US swine herd with human immunodeficiency virus infection: a double-
Intracellular transport of Toxoplasma gondii through the
using sera collected during the National Animal Health blind, randomized trial. J Infect Dis. 1996;173:91-97.
blood-brain barrier. J Neuroimmunol. 2011;232:119-130.
Monitoring Survey (Swine 2006). Zoonoses Public Health. 85. San-Andres FJ, Rubio R, Castilla J, et al. Incidence of
27. Carruthers V, Boothroyd JC. Pulling together: an inte-
2010;57:53-59. acquired immunodeficiency syndrome-associated oppor-
grated model of Toxoplasma cell invasion. Curr Opin
58. Dubey JP, Hill DE, Rozeboom DW, et al. High prevalence tunistic diseases and the effect of treatment on a cohort of
Microbiol. 2007;10:83-89.
and genotypes of Toxoplasma gondii isolated from organic 1115 patients infected with human immunodeficiency
28. Boothroyd JC, Dubremetz JF. Kiss and spit: the dual roles
pigs in northern USA. Vet Parasitol. 2012;188:14-18. virus, 1989-1997. Clin Infect Dis. 2003;36:1177-1185.
of Toxoplasma rhoptries. Nat Rev Microbiol. 2008;6:79-88.
59. Dubey JP, Gamble HR, Hill D, et al. High prevalence of 86. Luft BJ, Remington JS. Toxoplasmic encephalitis in AIDS
29. Conley FK, Jenkins KA, Remington JS. Toxoplasma gondii
viable Toxoplasma gondii infection in market weight pigs (AIDS commentary). Clin Infect Dis. 1992;15:211-222.
infection of the central nervous system. Use of the

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3153.e2
87. Abgrall S, Rabaud C, Costagliola D. Incidence and risk 114. Rosowski EE, Lu D, Julien L, et al. Strain-specific activation 140. Kelly MN, Kolls JK, Happel K, et al. Interleukin-17/
factors for toxoplasmic encephalitis in human immunode- of the NF-kappaB pathway by GRA15, a novel Toxoplasma interleukin-17 receptor-mediated signaling is important
ficiency virus-infected patients before and during the gondii dense granule protein. J Exp Med. 2011;208:195- for generation of an optimal polymorphonuclear response
highly active antiretroviral therapy era. Clin Infect Dis. 212. against Toxoplasma gondii infection. Infect Immun. 2005;
Part III  Infectious Diseases and Their Etiologic Agents

2001;33:1747-1755. 115. Yamamoto M, Ma JS, Mueller C, et al. ATF6beta is a host 73:617-621.


88. Jones JL, Sehgal M, Maguire JH. Toxoplasmosis-associated cellular target of the Toxoplasma gondii virulence factor 141. Lieberman LA, Cardillo F, Owyang AM, et al. IL-23 pro-
deaths among human immunodeficiency virus-infected ROP18. J Exp Med. 2011;208:1533-1546. vides a limited mechanism of resistance to acute toxo­
persons in the United States, 1992-1998. Clin Infect Dis. 116. Witola WH, Mui E, Hargrave A, et al. NALP1 influences plasmosis in the absence of IL-12. J Immunol. 2004;173:
2002;34:1161. susceptibility to human congenital toxoplasmosis, proin- 1887-1893.
89. Clumeck N. Some aspects of the epidemiology of toxoplas- flammatory cytokine response, and fate of Toxoplasma 142. Stumhofer JS, Laurence A, Wilson EH, et al. Interleukin
mosis and pneumocystosis in AIDS in Europe. Eur J Clin gondii-infected monocytic cells. Infect Immun. 2011;79: 27 negatively regulates the development of interleukin
Microbiol Infect Dis. 1991;10:177-178. 756-766. 17-producing T helper cells during chronic inflammation
90. Zumla A, Savva D, Wheeler RB, et al. Toxoplasma serology 117. Hunter CA, Subauste C, Van Cleave V, Remington JS. Pro- of the central nervous system. Nat Immunol. 2006;7:
in Zambian and Ugandan patients infected with the human duction of gamma interferon by natural killer cells from 937-945.
immunodeficiency virus. Trans R Soc Trop Med Hyg. 1991; Toxoplasma gondii-infected SCID mice: regulation by 143. Bohne W, Hessemann J, Gross U. Reduced replication of
85:227-229. interleukin-10, interleukin-12, and tumor necrosis factor Toxoplasma gondii is necessary for induction of bradyzoite-
91. Belanger F, Derouin F, Grangeot-Keros L, Meyer L. Inci- alpha. Infect Immun. 1994;62:2818-2824. specific antigens: a possible role for nitric oxide in trigger-
dence and risk factors of toxoplasmosis in a cohort of 118. Khan I, Matsuura T, Kasper L. Interleukin-12 enhances ing stage conversion. Infect Immun. 1994;62:1761-1767.
human immunodeficiency virus-infected patients: 1988- murine survival against acute toxoplasmosis. Infect Immun. 144. Chao C, Gekker G, Hu S, Peterson P. Human microglial
1995. HEMOCO and SEROCO Study Groups. Clin Infect 1994;62:1639-1642. cell defense against Toxoplasma gondii. J Immunol.
Dis. 1999;28:575-581. 119. Yap GS, Sher A. Cell-mediated immunity to Toxoplasma 1994;152:1246-1252.
92. Dankner WM, Lindsey JC, Levin MJ. Correlates of oppor- gondii: initiation, regulation and effector function. Immu- 145. Khan IA, Matssuura T, Fonseka S, Kasper LH. Production
tunistic infections in children infected with the human nobiology. 1999;201:240-247. of nitric oxide (NO) is not essential for protection against
immunodeficiency virus managed before highly active 120. Goldszmid RS, Caspar P, Rivollier A, et al. NK cell-derived acute Toxoplasma gondii infection in IRF-1−/− mice. J Immu-
antiretroviral therapy. Pediatr Infect Dis. 2001;20:40-48. interferon-gamma orchestrates cellular dynamics and the nol. 1996;156:636-643.
93. Lucas S, Hounnou A, Peacock C, et al. The mortality and differentiation of monocytes into dendritic cells at the site 146. Scharton-Kersten TM, Wynn TA, Denkers EY, et al. In the
pathology of HIV infection in a West African city. AIDS. of infection. Immunity. 2012;36:1047-1059. absence of endogenous IFN-γ mice develop unimpaired
1993;7:1569-1579. 121. Dunay IR, Fuchs A, Sibley LD. Inflammatory monocytes IL-12 reponses to Toxoplasma gondii while failing to
94. Wallace MR, Rossetti RJ, Olson PE. Cats and toxoplasmosis but not neutrophils are necessary to control infection with control acute infection. J Immunol. 1996;157:4045-4054.
risk in HIV-infected adults. JAMA. 1993;269:76-77. Toxoplasma gondii in mice. Infect Immun. 2010;78:1564- 147. Hayashi S, Chan C, Gazzinelli R, Roberge FG. Contribu-
95. Ruskin J, Remington JS. Toxoplasmosis in the compro- 1570. tion of nitric oxide to the host parasite equilibrium in toxo-
mised host. Ann Intern Med. 1976;84:193-199. 122. Subauste CS, Wessendarp M, Sorensen RU, Leiva LE. plasmosis. J Immunol. 1996;156:1476-1481.
96. Jones TC, Yeh S, Hirsch JG. The interaction between Toxo- CD40-CD40 ligand interaction is central to cell-mediated 148. Scharton-Kersten TM, Yap G, Magram J, Sher A. Inducible
plasma gondii and mammalian cells. I. Mechanism of entry immunity against Toxoplasma gondii: patients with hyper nitric oxide is essential for host control of persistent but
and intracellular fate of the parasite. J Exp Med. 1972;136: IgM syndrome have a defective type 1 immune response not acute infection with the intracellular pathogen Toxo-
1157-1172. that can be restored by soluble CD40 ligand trimer. plasma gondii. J Exp Med. 1997;185:1261-1273.
97. Murray HW, Nathans CF, Cohn ZA. Macrophages oxygen- J Immunol. 1999;162:6690-6700. 149. Taylor GA, Feng CG, Sher A. Control of IFN-gamma-
dependent antimicrobial activity. IV. Role of endogenous 123. Levy J, Espanol-Boren T, Thomas C, et al. Clinical spec- mediated host resistance to intracellular pathogens by
scavengers of oxygen intermediates. J Exp Med. 1980;152: trum of X-linked hyper-IgM syndrome. J Pediatr. 1997; immunity-related GTPases (p47 GTPases). Microbes Infect.
1610-1624. 131:47-54. 2007;9:1644-1651.
98. Sibley LD, Weidner E, Krahenbuhl JL. Phagosome acidifi- 124. Subauste CS, Wessendarp M, Smulian AG, Frame PT. Role 150. Martens S, Parvanova I, Zerrahn J, et al. Disruption of
cation blocked by intracellular Toxoplasma gondii. Nature. of CD40 ligand signaling in defective type 1 cytokine Toxoplasma gondii parasitophorous vacuoles by the mouse
1985;315:416-419. response in human immunodeficiency virus infection. p47-resistance GTPases. PLoS Pathog. 2005;1:e24.
99. Adams LB, Hibbs JB Jr, Taintor RR, Krahenbuhl JL. Micro- J Infect Dis. 2001;183:1722-1731. 151. Ling YM, Shaw MH, Ayala C, et al. Vacuolar and plasma
biostatic effect of murine-activated macrophages for Toxo- 125. Hunter CA, Remington JS. Immunopathogenesis of toxo- membrane stripping and autophagic elimination of Toxo-
plasma gondii. J Immunol. 1990;144:2725-2729. plasmic encephalitis. J Infect Dis. 1994;170:1057-1067. plasma gondii in primed effector macrophages. J Exp Med.
100. Pfefferkorn ER. Interferon gamma blocks the growth of 126. Gazzinelli R, Hieny S, Wynn T, et al. Interleukin 12 is 2006;203:2063-2071.
Toxoplasma gondii in human fibroblasts by inducing the required for the T-lymphocyte-independent induction of 152. Pavia CS. Protection against experimental toxoplasmosis
host cells to degrade tryptophan. Proc Natl Acad Sci U S A. interferon γ by an intracellular parasite and induces resis- by adoptive immunotherapy. J Immunol. 1986;137:2985-
1984;81:908-912. tance in T-cell-deficient hosts. Proc Natl Acad Sci U S A. 2990.
101. Schreiber RD, Feldman HA. Identification of the activator 1993;90:6115-6119. 153. Peterson P, Gekker G, Hu S, Chao C. Human astrocytes
system for antibody to Toxoplasma as the classical comple- 127. Gazzinelli R, Wysocka M, Hayashi S, et al. Parasite-induced inhibit intracellular multiplication of Toxoplasma gondii by
ment pathway. J Infect Dis. 1980;141:366-369. IL-12 stimulates early IFN-γ synthesis and resistance a nitric oxide-mediated mechanism. J Infect Dis. 1995;171:
102. Hunn JP, Feng CG, Sher A, Howard JC. The immunity- during acute infection with Toxoplasma gondii. J Immunol. 516-518.
related GTPases in mammals: a fast-evolving cell- 1994;153:2533-2543. 154. Brown CR, McLeod R. Class I MHC genes and CD8+ T
autonomous resistance system against intracellular 128. Suzuki Y, Orellana MA, Schreiber RD, Remington JS. cells determine cyst number in Toxoplasma gondii infec-
pathogens. Mamm Genome. 2011;22:43-54. Interferon-γ: the major mediator of resistance against Toxo- tion. J Immunol. 1990;145:3438-3441.
103. Degrandi D, Konermann C, Beuter-Gunia C, et al. Exten- plasma gondii. Science. 1988;240:516-518. 155. Brown C, Hunter C, Estes R, et al. Definitive identification
sive characterization of IFN-induced GTPases mGBP1 to 129. Suzuki Y, Conley FK, Remington JS. Importance of endog- of a gene that confers resistance against Toxoplasma cyst
mGBP10 involved in host defense. J Immunol. 2007;179: enous IFN-γ for prevention of toxoplasmic encephalitis in burden and encephalitis. Immunology. 1995;85:419-428.
7729-7740. mice. J Immunol. 1989;143:2045-2050. 156. Suzuki Y, Joh K, Orellna MA, et al. A gene(s) within the
104. Remington JS, Jacobs L, Kaufman HE. Studies on chronic 130. Suzuki Y, Conley FK, Remington JS. Treatment of toxoplas- H-2D region determines the development of toxoplasmic
toxoplasmosis: the relation of infective dose to residual mic encephalitis in mice with recombinant gamma inter- encephalitis in mice. Immunology. 1991;74:732-739.
infection and to the possiblity of congenital transmission. feron. Infect Immun. 1990;58:3050-3055. 157. Suzuki Y, Wong S-Y, Grumet FC, et al. Evidence for genetic
Am J Ophthalmol. 1958;46:261-267. 131. Robben PM, Mordue DG, Truscott SM, et al. Production regulation of susceptibility to toxoplasmic encephalitis in
105. Brown AS. Further evidence of infectious insults in the of IL-12 by macrophages infected with Toxoplasma gondii AIDS patients. J Infect Dis. 1996;173:265-268.
pathogenesis and pathophysiology of schizophrenia. Am J depends on the parasite genotype. J Immunol. 2004;172: 158. Mack D, Johnson J, Roberts F, et al. HLA-class II genes
Psychiatry. 2011;168:764-766. 3686-3694. modify outcome of Toxoplasma gondii infection. Int J Para-
106. Hofflin JM, Conley FK, Remington JS. Murine model of 132. Saeij JP, Coller S, Boyle JP, et al. Toxoplasma co-opts host sitol. 1999;29:1351-1358.
intracerebral toxoplasmosis. J Infect Dis. 1987;155:550-557. gene expression by injection of a polymorphic kinase 159. Habegger de Sorrentino A, Lopez R, Motta P, et al. HLA
107. Hofflin JM, Remington JS. Tissue culture isolation of Toxo- homologue. Nature. 2007;445:324-327. class II involvement in HIV-associated toxoplasmic enceph-
plasma from blood of a patient with AIDS. Arch Intern 133. Sibley LD, Adams LB, Fukutomi Y, Krahenbuhl JL. Tumor alitis development. Clin Immunol. 2005;115:133-137.
Med. 1985;145:925-926. necrosis factor-α triggers antitoxoplasmal activity of IFN- γ 160. Cordeiro CA, Moreira PR, Andrade MS, et al. Interleukin-10
108. Tirard V, Niel G, Rosenheim M, et al. Diagnosis of toxo- primed macrophages. J Immunol. 1991;147:2340-2345. gene polymorphism (-1082G/A) is associated with toxo-
plasmosis in patients with AIDS by isolation of the parasite 134. Langermans J, van der Hulst M, Nibbering P, et al. IFN- γ plasmic retinochoroiditis. Invest Ophthalmol Vis Sci. 2008;
from the blood. N Engl J Med. 1991;324:632. induced l-arginine-dependent toxoplasmastatic activity in 49:1979-1982.
109. Elbez-Rubinstein A, Ajzenberg D, Darde ML, et al. Con- murine peritoneal macrophages is mediated by endoge- 161. Jamieson SE, Peixoto-Rangel AL, Hargrave AC, et al. Evi-
genital toxoplasmosis and reinfection during pregnancy: nous tumor necrosis factor. J Immunol. 1992;148:568-574. dence for associations between the purinergic receptor
case report, strain characterization, experimental model of 135. Chang H, Pechere J, Piguet P. Role of tumour necrosis P2X(7) (P2RX7) and toxoplasmosis. Genes Immun. 2010;
reinfection, and review. J Infect Dis. 2009;199:280-285. factor in chronic murine Toxoplasma gondii encephalitis. 11:374-383.
110. Hunter CA, Sibley LD. Modulation of innate immunity by Immunol Infect Dis. 1992;2:61-68. 162. Peixoto-Rangel AL, Miller EN, Castellucci L, et al. Candi-
Toxoplasma gondii virulence effectors. Nat Rev Microbiol. 136. Suzuki Y, Yang Q, Yang S, et al. IL-4 is protective against date gene analysis of ocular toxoplasmosis in Brazil: evi-
2012;10:766-778. development of toxoplasmic encephalitis. J Immunol. dence for a role for toll-like receptor 9 (TLR9). Mem Inst
111. Denkers EY, Bzik DJ, Fox BA, Butcher BA. An inside job: 1996;157:2564-2569. Oswaldo Cruz. 2009;104:1187-1190.
hacking into Janus kinase/signal transducer and activator 137. Suzuki Y, Rani S, Liesenfeld O, et al. Impaired resistance to 163. Dorfman RF, Remington JS. Value of lymph-node biopsy
of transcription signaling cascades by the intracellular pro- the development of toxoplasmic encephalitis in interleukin- in the diagnosis of acute acquired toxoplasmosis. N Engl J
tozoan Toxoplasma gondii. Infect Immun. 2012;80:476-482. 6-defecient mice. Infect Immun. 1997;65:2339-2345. Med. 1973;289:878-881.
112. Reese ML, Boyle JP. Virulence without catalysis: how can a 138. Kasper LH, Matsuura T, Khan IA. IL-7 stimulates protec- 164. Montoya JG, Jordan R, Lingamneni S, et al. Toxoplasmic
pseudokinase affect host cell signaling? Trends Parasitol. tive immunity in mice against the intracellular pathogen, myocarditis and polymyositis in patients with acute
2012;28:53-57. Toxoplasma gondii. J Immunol. 1995;155:4798-4804. acquired toxoplasmosis diagnosed during life. Clin Infect
113. Howard JC, Hunn JP, Steinfeldt T. The IRG protein-based 139. Hunter CA, Neyer LA, Gabriel KE, et al. The role of CD28/ Dis. 1997;24:676-683.
resistance mechanism in mice and its relation to virulence B7 interaction in the regulation of NK cell responses 165. Negri Gualdi C, Negriaagualdi C. [Infectious pseudo-
in Toxoplasma gondii. Curr Opin Microbiol. 2011;14:414- during infection with Toxoplasma gondii. J Immunol. mononucleosis in lymphomatous diseases]. Minerva Med.
421. 1997;158:2285-2293. 1974;65:78-83 [in Italian].

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3153.e3
166. Weiss L, Chen Y, Berry G, et al. Infrequent detection of 193. Brion J-P, Pelloux H, Le Marc’hadour F, et al. Acute toxo- 223. Montoya JG, Giraldo LF, Efron B, et al. Infectious compli-
Toxoplasma gondii genome in toxoplasmic lymphadenitis: plasmic hepatitis in a patient with AIDS. Clin Infect Dis. cations among 620 consecutive heart transplant patients
a polymerase chain reaction study. Hum Pathol. 1992;23: 1992;15:183-184. at Stanford University Medical Center. Clin Infect Dis.
154-158. 194. Bergin C, Murphy M, Lyons D, et al. Toxoplasma pneumo- 2001;33:629-640.

Chapter 280  Toxoplasma gondii


167. Montoya JG, Remington JS. Toxoplasmosis of the central nitis: fatal presentation of disseminated toxoplasmosis in a 224. Wreghitt TG, Gray JJ, Pavel P, et al. Efficacy of pyrimeth-
nervous system. In: Peterson PK, Remington JS, eds. In patient with AIDS. Eur Respir J. 1992;5:1018-1020. amine for the prevention of donor-acquired Toxoplasma
Defense of the Brain: Current Concepts in the Immunopatho- 195. Crider SR, Horstman WG, Massey GS. Toxoplasma orchi- gondii infection in heart and heart-lung transplant patients.
genesis and Clinical Aspects of CNS Infections. Boston: tis: report of a case and a review of the literature. Am J Med. Transplant Int. 1992;5:197-200.
Blackwell Scientific; 1997:163-188. 1988;85:421-424. 225. Wreghitt TG, McNeil K, Roth C, et al. Antibiotic prophy-
168. Frenkel JK. Pathology and pathogenesis of congenital toxo- 196. Groll A, Schneider M, Althoff PH, et al. Morphology and laxis for the prevention of donor-acquired Toxoplasma
plasmosis. Bull N Y Acad Med. 1974;50:182-191. clinical significance of AIDS-related lesions in the adrenals gondii infection in transplant patients [letter; comment].
169. Ferguson DJ, Bowker C, Jeffery KJ, et al. Congenital toxo- and pituitary. Dtsch Med Wschr. 1990;115:483-488. J Infect. 1995;31:253-254.
plasmosis: continued parasite proliferation in the fetal 197. Patrick AL, Roberts LA, Burton EN, et al. Focal and seg- 226. Renoult E, Georges E, Biava MF, et al. Toxoplasmosis in
brain despite maternal immunological control in other mental glomerulosclerosis in the acquired immunodefi- kidney transplant recipients: report of six cases and review.
tissues. Clin Infect Dis. 2013;56:204-208. ciency syndrome. West Indian Med J. 1986;1986:200-202. Clin Infect Dis. 1997;24:625-634.
170. Wong B, Gold JWM, Brown AE, et al. Central-nervous- 198. Renold C, Sugar A, Chave J-P, et al. Toxoplasma encepha- 227. Filloy A, Garcia-Garcia O, Fernandez-Lorente L. Chorio-
system toxoplasmosis in homosexual men and parenteral litis in patients with the acquired immunodeficiency syn- retinitis as the first sign of acquired toxoplasmosis trans-
drug abusers. Ann Intern Med. 1984;100:36-42. drome. Medicine (Baltimore). 1992;71:224-239. mitted from donor following kidney transplantation: case
171. Post MJ, Chan JC, Hensley GT, et al. Toxoplasmosis 199. Remington JS. Toxoplasmosis in the adult. Bull N Y Acad report and review of the literature. Ocul Immunol Inflamm.
encephalitis in Haitian adults with acquired immunodefi- Med. 1974;50:211-227. 2013;21:27-28.
ciency syndrome: a clinical-pathologic-CT correlation. Am 200. Demar M, Ajzenberg D, Maubon D, et al. Fatal outbreak of 228. Assi MA, Rosenblatt JE, Marshall WF. Donor-transmitted
J Neuroradiol. 1983;4:155-162. human toxoplasmosis along the Maroni River: epidemio- toxoplasmosis in liver transplant recipients: a case report
172. Strittmatter C, Lang W, Wiestler OD, Kleihues P. The logical, clinical, and parasitological aspects. Clin Infect Dis. and literature review. Transpl Infect Dis. 2007;9:132-136.
changing pattern of human immunodeficiency virus asso- 2007;45:e88-e95. 229. Singer MA, Hagler WS, Grossniklaus HE. Toxoplasma
ciated cerebral toxoplasmosis: a study of 46 postmortem 201. Demar M, Hommel D, Djossou F, et al. Acute toxoplasmo- gondii retinochoroiditis after liver transplantation. Retina.
cases. Acta Neuropathol. 1992;83:475-481. ses in immunocompetent patients hospitalized in an inten- 1993;13:40-45.
173. Parmley SF, Goebel FD, Remington JS. Detection of sive care unit in French Guiana. Clin Microbiol Infect. 230. Botterel F, Ichai P, Feray C, et al. Disseminated toxoplas-
Toxoplasma gondi DNA in cerebrospinal fluid from AIDS 2012;18:E221-E231. mosis, resulting from infection of allograft, after orthotopic
patients by polymerase chain reaction. J Clin Microbiol. 202. McCabe RE, Brooks RG, Dorfman RF, Remington JS. liver transplantation: usefulness of quantitative PCR. J Clin
1992;30:3000-3002. Clinical spectrum in 107 cases of toxoplasmic lymphade- Microbiol. 2002;40:1648-1650.
174. Gray F, Gherardi R, Wingate E, et al. Diffuse “encephalitic” nopathy. Rev Infect Dis. 1987;9:754-774. 231. Martino R, Bretagne S, Rovira M, et al. Toxoplasmosis after
cerebral toxoplasmosis in AIDS. J Neurol. 1989;236:273- 203. Remington JS, Barnett CG, Meikel M, Lunde MN. Toxo- hematopoietic stem transplantation. Report of a 5-year
277. plasmosis and infectious mononucleosis. Arch Intern Med. survey from the Infectious Diseases Working Party of the
175. Mariuz P, Bosler EM, Luft BJ. Toxoplasma pneumonia. 1962;110:744-753. European Group for Blood and Marrow Transplantation.
Semin Respir Infect. 1997;12:40-43. 204. Wong WK, Upton A, Thomas MG. Neuropsychiatric Bone Marrow Transplant. 2000;25:1111-1114.
176. Bretagne S, Costa J-M, Fleury-Feith J, et al. Quantitative symptoms are common in immunocompetent adult 232. Martino R, Bretagne S, Einsele H, et al. Early detection of
competitive PCR with bronchoalveolar lavage fluid for patients with Toxoplasma gondii acute lymphadenitis. Toxoplasma infection by molecular monitoring of Toxo-
diagnosis of toxoplasmosis in AIDS patients. J Clin Micro- Scand J Infect Dis. 2013;45:357-361. plasma gondii in peripheral blood samples after allogeneic
biol. 1995;33:1662-1664. 205. Couvreur J, Thulliez P. [Acquired toxoplasmosis with stem cell transplantation. Clin Infect Dis. 2005;40:67-78.
177. Holland GN, O’Connor GR, Belfort R Jr, Remington JS. ocular or neurologic involvement]. Presse Med. 1996;25: 233. Matsuo Y, Takeishi S, Miyamoto T, et al. Toxoplasmosis
Toxoplasmosis. In: Pepose JS, Holland GN, Wilhelmus KR, 438-442 [in French]. encephalitis following severe graft-vs.-host disease after
eds. Ocular Infection and Immunity. St Louis: Mosby Year- 206. Montoya JG, Remington JS. Toxoplasmic chorioretinitis in allogeneic hematopoietic stem cell transplantation: 17 yr
book; 1996:1183-1223. the setting of acute acquired toxoplasmosis. Clin Infect Dis. experience in Fukuoka BMT group. Eur J Haematol. 2007;
178. Danise A, Cinque P, Vergani S, et al. Use of the polymerase 1996;23:277-282. 79:317-321.
chain reaction assays of aqueous humor in the differential 207. Delair E, Monnet D, Grabar S, et al. Respective roles of 234. Caner A, Donmez A, Doskaya M, et al. Determining Toxo-
diagnosis of retinitis in patients infected with human acquired and congenital infections in presumed ocular plasma high-risk autologous and allogeneic hematopoietic
immunodeficiency virus. Clin Infect Dis. 1997;24:1100- toxoplasmosis. Am J Ophthalmol. 2008;146:851-855. stem cell transplantation patients by systematic pre-
1106. 208. Burnett AJ, Shortt SG, Isaac-Renton J, et al. Multiple cases transplant PCR screening of stem cell originated buffy coat.
179. Montoya JG, Parmley S, Liesenfeld O, et al. Use of the of acquired toxoplasmosis retinitis presenting in an out- Parasitology Int. 2012;61:565-571.
polymerase chain reaction for diagnosis of ocular toxoplas- break. Ophthalmology. 1998;105:1032-1037. 235. Sing A, Leitritz L, Roggenkamp A, et al. Pulmonary toxo-
mosis. Ophthalmology. 1999;106:1554-1563. 209. Toporovski J, Romano S, Hartmann S, et al. Nephrotic plasmosis in bone marrow transplant recipients: report of
180. Silveira C, Belfort R Jr, Muccioli C, et al. The effect of long- syndrome associated with toxoplasmosis: report of seven two cases and review. Clin Infect Dis. 1999;29:429-433.
term intermittent trimethoprim/sulfamethoxazole treat- cases. Rev Inst Med Trop São Paulo. 2012;54:61-64. 236. Liesenfeld O, Wong SY, Remington JS. Toxoplasmosis in
ment on recurrences of toxoplasmic retinochoroiditis. Am 210. Montoya JG, Remington JS. Studies on the serodiagnosis the setting of AIDS. In: Bartlett JG, Merigan TC, Bolognesi
J Ophthalmol. 2002;134:41-46. of toxoplasmic lymphadenitis. Clin Infect Dis. 1995;20:781- D, eds. Textbook of AIDS Medicine. 2nd ed. Baltimore:
181. Jones JL, Muccioli C, Belfort R Jr, et al. Recently acquired 790. Williams & Wilkins; 1999:225-259.
Toxoplasma gondii infection, Brazil. Emerg Infect Dis. 2006; 211. Cunningham T. Pancarditis in acute toxoplasmosis. Am J 237. Oksenhendler E, Cadranel J, Sarfati C, et al. Toxoplasma
12:582-587. Clin Pathol. 1982;78:403-405. gondii pneumonia in patients with the acquired immuno-
182. Gilbert RE, Freeman K, Lago EG, et al. Ocular sequelae of 212. Prado SP, Pacheco VC, Noemi IH, et al. [Toxoplasma peri- deficiency syndrome. Am J Med. 1990;88(5N):18N-21N.
congenital toxoplasmosis in Brazil compared with Europe. carditis and myocarditis]. Rev Chil Pediatr. 1978;49:179- 238. Mehren M, Burns PJ, Mamani F, et al. Toxoplasmic myelitis
PLoS Negl Trop Dis. 2008;2:e277. 185 [in Spanish]. mimicking intramedullary spinal cord tumor. Neurology.
183. de-la-Torre A, Lopez-Castillo CA, Gomez-Marin JE. Inci- 213. Greenlee JE, Johnson WD Jr, Campa JF, et al. Adult toxo- 1988;38:1648-1650.
dence and clinical characteristics in a Colombian cohort of plasmosis presenting as polymyositis and cerebellar ataxia. 239. Herskovitz S, Siegel SE, Schneider AT, et al. Spinal cord
ocular toxoplasmosis. Eye. 2009;23:1090-1093. Ann Intern Med. 1975;82:367-371. toxoplasmosis in AIDS. Neurology. 1989;39:1552-1553.
184. Vallochi AL, Muccioli C, Martins MC, et al. The genotype 214. Paspalaki PK, Mihailidou EP, Bitsori M, et al. Polymyositis 240. Overhage JM, Greist A, Brown DR. Conus medullaris syn-
of Toxoplasma gondii strains causing ocular toxoplasmo- and myocarditis associated with acquired toxoplasmosis drome resulting from Toxoplasma gondii infection in a
sis in humans in Brazil. Am J Ophthalmol. 2005;139: in an immunocompetent girl. BMC Musculoskelet Disord. patient with the acquired immunodeficiency syndrome.
350-351. 2001;2:8. Am J Med. 1990;89:814-815.
185. Grigg ME, Ganatra J, Boothroyd JC, Margolis TP. Unusual 215. Palma S, Reyes H, Guzman L, Cartier L. [Dermatomyositis 241. Rabaud C, May T, Lucet JC, et al. Pulmonary toxoplasmosis
abundance of atypical strains associated with human and toxoplasmosis]. Rev Med Chil. 1984;111:164-167 [in in patients infected with human immunodeficiency virus:
ocular toxoplasmosis. J Infect Dis. 2001;184:633-639. Spanish]. a French national study. Clin Infect Dis. 1996;23:1249-
186. Gherardi R, Baudrimont M, Lionnet F, et al. Skeletal 216. Pollock JL. Toxoplasmosis appearing to be dermatomyosi- 1254.
muscle toxoplasmosis in patients with acuqired immuno- tis. Arch Dermatol. 1979;115:736-737. 242. Derouin F, Sarfati C, Beauvais B, et al. Prevalence of pul-
deficiency syndrome: a clinical and pathological study. Ann 217. Brown AS, Schaefer CA, Quesenberry CP Jr, et al. Maternal monary toxoplasmosis in HIV-infected patients. AIDS.
Neurol. 1992;32:535-542. exposure to toxoplasmosis and risk of schizophrenia in 1990;4:1036.
187. Calico I, Caballero E, Martinez O, et al. Isolation of Toxo- adult offspring. Am J Psychiatry. 2005;162:767-773. 243. Holland G, Engstrom R Jr, Glasgow B, et al. Ocular toxplas-
plasma gondii from immunocompromised patients using 218. Niebuhr DW, Millikan AM, Cowan DN, et al. Selected mosis in patients with acquired immunodeficiency syn-
tissue culture. Infection. 1991;19:340-342. infectious agents and risk of schizophrenia among U.S. drome. Am J Ophtalmol. 1988;106:653-667.
188. Roldan EO, Moskowitz L, Hensley GT. Pathology of the military personnel. Am J Psychiatry. 2008;165:99-106. 244. Heinemann MH, Gold JM, Maisel J. Bilateral Toxoplasma
heart in acquired immunodeficiency syndrome. Arch 219. Torrey EF, Bartko JJ, Yolken RH. Toxoplasma gondii and retinochoroiditis in a patient with acquired immune defi-
Pathol Lab Med. 1987;111:943-946. other risk factors for schizophrenia: an update. Schizophr ciency syndrome. Retina. 1986;6:224-227.
189. Adair OV, Randive N, Krasnow N. Isolated Toxoplasma Bull. 2012;38:642-647. 245. Louie JK, Hsu LC, Osmond DH, et al. Trends in causes of
myocarditis in acquired immune deficiency syndrome. Am 220. Lassoued S, Zabraniecki L, Marin F, Billey T. Toxoplasmic death among persons with acquired immunodeficiency
Heart J. 1989;118:856-857. chorioretinitis and antitumor necrosis factor treatment in syndrome in the era of highly active antiretroviral therapy,
190. Pauwels A, Meyohas MC, Eliaszewicz M, et al. Toxoplasma rheumatoid arthritis. Semin Arthritis Rheum. 2007;36:262- San Francisco, 1994-1998. J Infect Dis. 2002;186:1023-
colitis in the acquired immunodeficiency syndrome. Am J 263. 1027.
Gastroenterol. 1992;87:518-519. 221. Luft BJ, Naot Y, Araujo FG, et al. Primary and reactivated 246. Antinori A, Larussa D, Cingolani A, et al. Prevalence, asso-
191. Smart PE, Weinfeld A, Thompson NE, Defortuna SM. Toxoplasma infection in patients with cardiac transplants. ciated factors, and prognostic determinants of AIDS-
Toxoplasmosis of the stomach: a cause of antral narrowing. Clinical spectrum and problems in diagnosis in a defined related toxoplasmic encephalitis in the era of advanced
Radiology. 1990;174:369-370. population. Ann Intern Med. 1983;99:27-31. highly active antiretroviral therapy. Clin Infect Dis. 2004;
192. Garcia LW, Hemphill RB, Marasco WA, Ciano PS. Acquired 222. Fernandez-Sabe N, Cervera C, Farinas MC, et al. Risk 39:1681-1691.
immunodeficiency syndrome with disseminated toxoplas- factors, clinical features, and outcomes of toxoplasmosis in 247. Ghosn J, Paris L, Ajzenberg D, et al. Atypical toxoplasmic
mosis presenting as an acute pulmonary and gastro- solid-organ transplant recipients: a matched case-control manifestation after discontinuation of maintenance
intestinal illness. Arch Pathol Lab Med. 1991;115:459-463. study. Clin Infect Dis. 2012;54:355-361. therapy in a human immunodeficiency virus type

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3153.e4
1-infected patient with immune recovery. Clin Infect Dis. 273. Desmonts G, Couvreur J. Congenital toxoplasmosis. A 298. Liesenfeld O, Montoya JG, Kinney S, et al. Effect of testing
2003;37:e112-e114. prospective study of the offspring of 542 women who for IgG avidity in the diagnosis of Toxoplasma gondii infec-
248. Sendi P, Sachers F, Drechsler H, Graber P. Immune recov- acquired toxoplasmosis during pregnancy. In: Thalhammer tion in pregnant women: experience in a US reference labo-
ery vitritis in an HIV patient with isolated toxoplasmic O, Pollak A, Baumgarten K, eds. Pathophysiology of Con- ratory. J Infect Dis. 2001;183:1248-1253.
Part III  Infectious Diseases and Their Etiologic Agents

retinochoroiditis. AIDS. 2006;20:2237-2238. genital Disease. Perinatal Medicine (6th European Congress, 299. Liesenfeld O, Montoya JG, Tathineni NJ, et al. Confirma-
249. Martin-Blondel G, Alvarez M, Delobel P, et al. Toxoplasmic Vienna). Stuttgart: Georg Thieme Publishers; 1979:51-60. tory serologic testing for acute toxoplasmosis and rate of
encephalitis IRIS in HIV-infected patients: a case series and 274. Koppe JG, Loewer-Sieger DH, De Roever-Bonnet H. induced abortions among women reported to have positive
review of the literature. J Neurol Neurosurg Psychiatry. Results of 20-year follow-up of congenital toxoplasmosis. Toxoplasma immunoglobulin M antibody titers. Am J
2011;82:691-693. Am J Ophthalmol. 1986;101:248-249. Obstet Gynecol. 2001;184:140-145.
250. Pfeffer G, Prout A, Hooge J, Maguire J. Biopsy-proven 275. Wilson CB, Remington JS, Stagno S, Reynolds DW. Devel- 300. Montoya JG, Liesenfeld O, Kinney S, et al. VIDAS test
immune reconstitution syndrome in a patient with AIDS opment of adverse sequelae in children born with subclini- for avidity of Toxoplasma-specific immunoglobulin G for
and cerebral toxoplasmosis. Neurology. 2009;73:321-322. cal congenital Toxoplasma infection. Pediatrics. 1980;66: confirmatory testing of pregnant women. J Clin Microbiol.
251. Furrer H, Opravil M, Bernasconi E, et al. Stopping primary 767-774. 2002;40:2504-2508.
prophylaxis in HIV-1-infected patients at high risk of Toxo- 276. Delhaes L, Ajzenberg D, Sicot B, et al. Severe congenital 301. Sabin AB, Feldman HA. Dyes as microchemical indicators
plasma encephalitis. Swiss HIV Cohort Study. Lancet. toxoplasmosis due to a Toxoplasma gondii strain with an of a new immunity phenomenon affecting a protozoan
2000;355:2217-2218. atypical genotype: case report and review. Prenat Diagn. parasite (Toxoplasma). Science. 1948;108:660-663.
252. Kirk O, Reiss P, Uberti-Foppa C, et al. Safe interruption of 2010;30:902-905. 302. Balsari A, Poli G, Molina V, et al. ELISA for Toxoplasma
maintenance therapy against previous infection with four 277. Forestier F. [Fetal diseases, prenatal diagnoses and practical antibody detection: a comparison with other serodiagnos-
common HIV-associated opportunistic pathogens during measures]. Presse Med. 1991;20:1448-1454 [in French]. tic tests. J Clin Pathol. 1980;33:640-643.
potent antiretroviral therapy. Ann Intern Med. 2002;137: 278. Hohlfeld P, Daffos F, Thulliez P, et al. Fetal toxoplasmosis: 303. Walls KW, Bullock SL, English DK. Use of the enzyme-
239-250. outcome of pregnancy and infant follow-up after in utero linked immunosorbent assay (ELISA) and its micro­
253. Miro JM, Lopez JC, Podzamczer D, et al. Discontinuation treatment. J Pediatr. 1989;115:765-769. adaptation for the serodiagnosis of toxoplasmosis. J Clin
of primary and secondary Toxoplasma gondii prophylaxis 279. Hohlfeld P, Daffos F, Costa J-M, et al. Prenatal diagnosis of Microbiol. 1977;5:273-277.
is safe in HIV-infected patients after immunological resto- congenital toxoplasmosis with polymerase-chain-reaction 304. Walton BC, Benchoff BM, Brooks WH. Comparison of the
ration with highly active antiretroviral therapy: results of test on amniotic fluid. N Engl J Med. 1994;331:695-699. indirect fluorescent antibody test and methylene blue dye
an open, randomized, multicenter clinical trial. Clin Infect 280. Thiebaut R, Leproust S, Chene G, Gilbert R. Effectiveness test for detection of antibodies to Toxoplasma gondii. Am J
Dis. 2006;43:79-89. of prenatal treatment for congenital toxoplasmosis: a meta- Trop Med Hyg. 1966;15:149-152.
254. Stout JE, Lai JC, Giner J, Hamilton CD. Reactivation of analysis of individual patients’ data. Lancet. 2007;369: 305. Thulliez P, Remington JS, Santoro F, et al. A new agglutina-
retinal toxoplasmosis despite evidence of immune response 115-122. tion test for the diagnosis of acute and chronic Toxoplasma
to highly active antiretroviral therapy. Clin Infect Dis. 281. Dunn D, Wallon M, Peyron F, et al. Mother-to-child trans- infection. Pathol Biol (Paris). 1986;34:173-177.
2002;35:e37-e39. mission of toxoplasmosis: risk estimates for clinical coun- 306. Araujo FG, Barnett EV, Gentry LO, Remington JS. False-
255. Khan A, Su C, German M, et al. Genotyping of Toxoplasma selling. Lancet. 1999;353:1829-1833. positive anti-Toxoplasma fluorescent-antibody tests in
gondii strains from immunocompromised patients reveals 282. Hotop A, Hlobil H, Gross U. Efficacy of rapid treatment patients with antinuclear antibodies. Appl Microbiol. 1971;
high prevalence of type I strains. J Clin Microbiol. 2005; initiation following primary Toxoplasma gondii infection 22:270-275.
43:5881-5887. during pregnancy. Clin Infect Dis. 2012;54:1545-1552. 307. Desmonts G, Remington JS. Direct agglutination test for
256. Holland GN. Reconsidering the pathogenesis of ocular 283. Cortina-Borja M, Tan HK, Wallon M, et al. Prenatal treat- diagnosis of Toxoplasma infection: method for increasing
toxoplasmosis. Am J Ophthalmol. 1999;128:502-505. ment for serious neurological sequelae of congenital toxo- sensitivity and specificity. J Clin Microbiol. 1980;11:562-
257. London NJ, Hovakimyan A, Cubillan LD, et al. Prevalence, plasmosis: an observational prospective cohort study. PLoS 568.
clinical characteristics, and causes of vision loss in patients Med. 2010;7. pii:e1000351. 308. Dannemann BR, Vaughan WC, Thulliez P, Remington JS.
with ocular toxoplasmosis. Eur J Ophthalmol. 2011;21: 284. Olariu TR, Remington JS, McLeod R, et al. Severe congeni- Differential agglutination test for diagnosis of recently
811-819. tal toxoplasmosis in the United States: clinical and sero- acquired infection with Toxoplasma gondii. J Clin Micro-
258. Silveira C, Belfort R Jr, Burnier M Jr, Nussenblatt R. logic findings in untreated infants. Pediatr Infect Dis J. biol. 1990;28:1928-1933.
Acquired toxoplasmic infection as the cause of toxoplasmic 2011;30:1056-1061. 309. Montoya JG, Berry A, Rosso F, Remington JS. The differ-
retinochoroiditis in families. Am J Ophthalmol. 1988;106: 285. Dubey JP, Lago EG, Gennari SM, et al. Toxoplasmosis in ential agglutination test as a diagnostic aid in cases of
362-364. humans and animals in Brazil: high prevalence, high toxoplasmic lymphadenitis. J Clin Microbiol. 2007;45:
259. Glasner PD, Silveira C, Kruszon-Moran D, et al. An unus- burden of disease, and epidemiology. Parasitology. 2012; 1463-1468.
ally high prevalence of ocular toxoplasmosis in Southern 139:1375-1424. 310. Hedman K, Lappalainen M, Seppala I, Makela O. Recent
Brazil. Am J Ophthalmol. 1992;114:136-144. 286. Romand S, Chosson M, Franck J, et al. Usefulness of quan- primary Toxoplasma infection indicated by a low avidity of
260. Silveira C, Belfort R Jr., Muccioli C, et al. A follow-up study titative polymerase chain reaction in amniotic fluid as early specific IgG. J Infect Dis. 1989;159:736-739.
of Toxoplasma gondii infection in southern Brazil. Am J prognostic marker of fetal infection with Toxoplasma 311. Lappalainen M, Koskela P, Koskiniemi M, et al. Toxoplas-
Ophthalmol. 2001;131:351-354. gondii. Am J Obstet Gynecol. 2004;190:797-802. mosis acquired during pregnancy: improved serodiagnosis
261. Nussenblatt R, Belfort R Jr. Ocular toxoplasmosis. JAMA. 287. Couvreur J, Desmonts G, Tournier G, Szusterkac M. [A based on avidity of IgG. J Infect Dis. 1993;167:691-697.
1994;271:302-307. homogeneous series of 210 cases of congenital toxoplasmo- 312. Montoya JG, Huffman HB, Remington JS. Evaluation of
262. Bosch-Driessen LE, Berendschot TT, Ongkosuwito JV, sis in 0 to 11-month-old infants detected prospectively]. the immunoglobulin G avidity test for diagnosis of toxo-
Rothova A. Ocular toxoplasmosis: clinical features and Ann Pediatr (Paris). 1984;31:815-819 [in French]. plasmic lymphadenopathy. J Clin Microbiol. 2004;42:4627-
prognosis of 154 patients. Ophthalmology. 2002;109:869- 288. De Roever-Bonnet H, Koppe JG, Loewer-Sieger DH. 4631.
878. Follow-up of children with congenital Toxoplasma infec- 313. Pelloux H, Brun E, Vernet G, et al. Determination of anti-
263. Bosch-Driessen LH, Plaisier MB, Stilma JS, et al. Reactiva- tion and children who become serologically negative after Toxoplasma gondii immunoglobulin G avidity: adaptation
tions of ocular toxoplasmosis after cataract extraction. 1 year of age, all born in 1964-1965. In: Thalhammer O, to the Vidas system (bioMérieux). Diagn Microbiol Infect
Ophthalmology. 2002;109:41-45. Pollak A, Baumgarten K, eds. Pathophysiology of Congenital Dis. 1998;32:69-73.
264. Holland GN, Crespi CM, ten Dam-van Loon N, et al. Disease. Perinatal Medicine (6th European Congress, 314. Villard O, Breit L, Cimon B, et al. Comparison of four
Analysis of recurrence patterns associated with toxoplas- Vienna). Stuttgart: Georg Thieme Publishers; 1979: commercially available avidity tests for Toxoplasma gondii-
mic retinochoroiditis. Am J Ophthalmol. 2008;145:1007- 61-75. specific IgG antibodies. Clin Vaccine Immunol. 2013;20:
1013. 289. Faucher B, Garcia-Meric P, Franck J, et al. Long-term 197-204.
264a.  Jones JL, Bonetti V, Holllan GN, et al. Ocular toxoplasmo- ocular outcome in congenital toxoplasmosis: a prospective 315. Bobic B, Sibalic D, Djurkovic-Djakovic O. High levels of
sis in the United States: recent and remote infections. Clin cohort of treated children. J Infect. 2012;64:104-109. IgM antibodies specific for Toxoplasma gondii in pregnancy
Infect Dis. 2015;60:271-273. 290. Berrebi A, Assouline C, Bessieres MH, et al. Long-term 12 years after primary Toxoplasma infection. Gynecol
265. Delair E, Latkany P, Noble AG, et al. Clinical manifesta- outcome of children with congenital toxoplasmosis. Am J Obstet Invest. 1991;31:182-184.
tions of ocular toxoplasmosis. Ocul Immunol Inflamm. Obstet Gynecol. 2010;203:552 e1-552 e6. 316. Liesenfeld O, Press C, Montoya JG, et al. False-positive
2011;19:91-102. 291. Phan L, Kasza K, Jalbrzikowski J, et al. Longitudinal study results in immunoglobulin M (IgM) Toxoplasma antibody
266. Johnson MW, Greven CM, Jaffe GJ, et al. Atypical, severe of new eye lesions in children with toxoplasmosis who were tests and importance of confirmatory testing: the Platelia
toxoplasmic retinochoroiditis in elderly patients. Ophthal- not treated during the first year of life. Am J Ophthalmol. toxo IgM test. J Clin Microbiol. 1997;35:174-178.
mology. 1997;104:48-57. 2008;146:375-384. 317. Wilson M, Ware DA, Walls KW. Evaluation of commercial
267. Kong JT, Grigg ME, Uyetake L, et al. Serotyping of Toxo- 292. Phan L, Kasza K, Jalbrzikowski J, et al. Longitudinal study serodiagnostic kits for toxoplasmosis. J Clin Microbiol.
plasma gondii infections in humans using synthetic pep- of new eye lesions in treated congenital toxoplasmosis. 1987;25:2262-2265.
tides. J Infect Dis. 2003;187:1484-1495. Ophthalmology. 2008;115:553-559.e8. 318. U.S. Food and Drug Administration. Public Health Advi-
268. Boyer K, Hill D, Mui E, et al. Unrecognized ingestion of 293. Mitchell CD, Erlich SS, Mastrucci MT, et al. Congenital sory: Center for Devices and Radiological Health. Limitations
Toxoplasma gondii oocysts leads to congenital toxoplasmo- toxoplasmosis occurring in infants perinatally infected of Toxoplasma IgM commercial test kits, July 25, 1997.
sis and causes epidemics in North America. Clin Infect Dis. with human immunodeficiency virus 1. Pediatr Infect Dis Rockville, MD: U.S. Food and Drug Administration; 1997:
2011;53:1081-1089. J. 1990;9:512-518. 1-3. Available at http://wwwfdagov/cdrh/toxophahtml.
269. Gavinet MF, Robert F, Firtion G, et al. Congenital toxoplas- 294. Nowakowska D, Colon I, Remington JS, et al. Genotyping 319. Montoya JG, Remington JS. Management of Toxoplasma
mosis due to maternal reinfection during pregnancy. J Clin of Toxoplasma gondii by multiplex PCR and peptide-based gondii infection during pregnancy. Clin Infect Dis. 2008;47:
Microbiol. 1997;35:1276-1277. serological testing of samples from infants in Poland diag- 554-566.
270. Hennequin C, Dureau P, N’Guyen L, et al. Congenital toxo- nosed with congenital toxoplasmosis. J Clin Microbiol. 320. Welch PC, Masur H, Jones TC, Remington JS. Serologic
plasmosis acquired from an immune woman. Pediatr Infect 2006;44:1382-1389. diagnosis of acute lymphadenopathic toxoplasmosis.
Dis J. 1997;16:75-76. 295. McLeod R, Boyer KM, Lee D, et al. Prematurity and sever- J Infect Dis. 1980;142:256-264.
271. Vogel N, Kirisits M, Michael E, et al. Congenital toxoplas- ity are associated with Toxoplasma gondii alleles (NCCCTS, 321. Naot Y, Barnett EV, Remington JS. Method for avoiding
mosis transmitted from an immunologically competent 1981-2009). Clin Infect Dis. 2012;54:1595-1605. false-positive results occurring in immunoglobulin M
mother infected before conception. Clin Infect Dis. 1996; 296. Montoya JG. Laboratory diagnosis of Toxoplasma gondii enzyme-linked immunosorbent assays due to presence of
23:1055-1060. infection and toxoplasmosis. J Infect Dis. 2002;185:S73- both rheumatoid factor and antinuclear antibodies. J Clin
272. Minkoff H, Remington JS, Holman S, et al. Vertical trans- S82. Microbiol. 1981;14:73-78.
mission of Toxoplasma by human immunodeficiency 297. Miller MJ, Aronson WJ, Remington JS. Late parasitemia in 322. Filice GA, Yeager AS, Remington JS. Diagnostic signifi-
virus-infected women. Am J Obstet Gynecol. 1997;176: asymptomatic acquired toxoplasmosis. Ann Intern Med. cance of immunoglobulin M antibodies to Toxoplasma
555-559. 1969;71:139-145. gondii detected after separation of immunoglobulin M

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3153.e5
from immunoglobulin G antibodies. J Clin Microbiol. 347. Jones CD, Okhravi N, Adamson P, et al. Comparison of 371. Giancola ML, Rizzi EB, Schiavo R, et al. Reduced value of
1980;12:336-342. PCR detection methods for B1, P30, and 18S rDNA genes thallium-201 single-photon emission computed tomogra-
323. Naot Y, Remington JS. An enzyme-linked immunosorbent of T. gondii in aqueous humor. Invest Ophthalmol Vis Sci. phy in the management of HIV-related focal brain lesions
assay for detection of IgM antibodies to Toxoplasma gondii: 2000;41:634-644. in the era of highly active antiretroviral therapy. AIDS Res

Chapter 280  Toxoplasma gondii


use for diagnosis of acute acquired toxoplasmosis. J Infect 348. Costa JM, Bretagne S. Variation of B1 gene and AF146527 Hum Retroviruses. 2004;20:584-588.
Dis. 1980;142:757-766. repeat element copy numbers according to Toxoplasma 372. Navia BA, Petito CK, Gold JW, et al. Cerebral toxoplasmo-
324. Naot Y, Desmonts G, Remington JS. IgM enzyme-linked gondii strains assessed using real-time quantitative PCR. sis complicating the acquired immune deficiency syn-
immunosorbent assay test for the diagnosis of congenital J Clin Microbiol. 2012;50:1452-1454. drome: clinical and neuropathological findings in 27
Toxoplasma infection. J Pediatr. 1981;98:32-36. 349. Kasper DC, Sadeghi K, Prusa AR, et al. Quantitative real- patients. Ann Neurol. 1986;19:224-238.
325. Siegel JP, Remington JS. Comparison of methods for quan- time polymerase chain reaction for the accurate detection 373. Orefice G, Carrieri PB, de Marinis T, et al. Use of the
titating antigen-specific immunoglobulin M antibody with of Toxoplasma gondii in amniotic fluid. Diagn Microbiol intrathecal synthesis of antitoxoplasma antibodies in the
a reverse enzyme-linked immunosorbent assay. J Clin Infect Dis. 2009;63:10-15. diagnositc assessment and in the follow-up of AIDS
Microbiol. 1983;18:63-70. 350. Morelle C, Varlet-Marie E, Brenier-Pinchart MP, et al. patients with cerebral toxoplasmosis. Acta Neurol (Napoli).
326. Remington JS, Eimstad WM, Araujo FG. Detection of Comparative assessment of a commercial kit and two 1990;12:79-81.
immunoglobulin M antibodies with antigen-tagged latex laboratory-developed PCR assays for the molecular diag- 374. Potasman I, Resnick L, Luft BJ, Remington JS. Intrathecal
particles in an immunosorbent assay. J Clin Microbiol. nosis of congenital toxoplasmosis. J Clin Microbiol. 2012; production of antibodies against Toxoplasma gondii in
1983;17:939-941. 50:3977-3982. patients with toxoplasmic encephalitis and the acquired
327. Plantaz D, Goullier A, Jouk PS, Bost M. [Value of the 350a.  Robert-Gangneux F, Sterkers Y, Yera H, et al. Molecular immunodeficiency syndrome (AIDS). Ann Intern Med.
immunosorbent agglutination assay (ISAGA) in the early diagnosis of toxoplasmosis in immunocompromised 1988;108:49-51.
diagnosis of congenital toxoplasmosis]. Pediatrie. 1987;42: patients: a three-year multicenter retrospective study. J Clin 375. Fricker-Hidalgo H, Bulabois CE, Brenier-Pinchart MP,
387-391 [in French]. Microbiol. 2015;53:1677-1684. et al. Diagnosis of toxoplasmosis after allogeneic stem cell
328. Decoster A, Slizewicz B, Simon J, et al. Platelia-toxo IgA, a 350b.  Meers S, Lagrou K, Theunissen K, et al. Myeloablative transplantation: results of DNA detection and serological
new kit for early diagnosis of congenital toxoplasmosis by conditioning predisposes patients for Toxoplasma gondii techniques. Clin Infect Dis. 2009;48:e9-e15.
detection of anti-P30 immunoglobulin A antibodies. J Clin reactivation after allogeneic stem cell transplantation. Clin 376. Deleted in proofs.
Microbiol. 1991;29:2291-2295. Infect Dis. 2010;50:1127-1134. 377. Bossolasco S, Cinque P, Ponzoni M, et al. Epstein-Barr
329. Pinon JM, Thoannes H, Pouletty PH, et al. Detection of IgA 351. Levy RM, Bredesen DE, Rosenblum ML. Opportunistic virus DNA load in cerebrospinal fluid and plasma of
specific for toxoplasmosis in serum and cerebrospinal fluid central nervous system pathology in patients with AIDS. patients with AIDS-related lymphoma. J Neurovirol. 2002;
using a non-enzymatic IgA-capture assay. Diagn Immunol. Ann Neurol. 1988;23:S7-S12. 8:432-438.
1986;4:223-227. 352. Frenkel JK, Piekarski G. The demonstration of Toxoplasma 378. Holliman RE, Johnson JD, Savva D. Diagnosis of cerebral
330. Stepick-Biek P, Thulliez P, Araujo FG, Remington JS. IgA and other organisms by immunofluorescence: a pitfall [edi- toxoplasmosis in association with AIDS using the poly-
antibodies for diagnosis of acute congenital and acquired torial]. J Infect Dis. 1978;138:265-266. merase chain reaction. Scand J Infect Dis. 1990;22:243-
toxoplasmosis. J Infect Dis. 1990;162:270-273. 353. Sun T, Greenspan J, Tenenbaum M, et al. Diagnosis of 244.
331. Pinon JM, Toubas D, Marx C, et al. Detection of specific cerebral toxoplasmosis using fluorescein-labeled antitoxo- 379. Roth A, Roth B, Höffken G, et al. Application of the poly-
immunoglobulin E in patients with toxoplasmosis. J Clin plasma monoclonal antibodies. Am J Surg Pathol. 1986;10: merase chain reaction to diagnosis of pulmonary toxo­
Microbiol. 1990;28:1739-1743. 312-316. plasmosis in immunocompromised patients. Eur J Clin
332. Wong SY, Hadju M-P, Ramirez R, et al. The role of specific 354. Cerezo L, Alvarez M, Price G. Electron microscopic diag- Microbiol Infect Dis. 1992;11:1177-1181.
immunoglobulin E in diagnosis of acute Toxoplasma infec- nosis of cerebral toxoplasmosis. Case report. J Neurosurg. 380. Fardeau C, Romand S, Rao NA, et al. Diagnosis of toxo-
tion and toxoplasmosis. J Clin Microbiol. 1993;31:2952- 1985;63:470-472. plasmic retinochoroiditis with atypical clinical features.
2959. 355. Luft BJ, Billingham M, Remington JS. Endomyocardial Am J Ophthalmol. 2002;134:196-203.
333. Poirriez J, Toubas D, Marx-Chemia C, et al. Isotypic char- biopsy in the diagnosis of toxoplasmic myocarditis. Trans- 381. Westeneng AC, Rothova A, de Boer JH, de Groot-Mijnes
acterization of anti-Toxoplasma gondii antibodies in 18 plant Proc. 1986;18:1871-1873. JD. Infectious uveitis in immunocompromised patients
cases of congenital toxoplasmic chorioretinitis. Acta Oph- 356. Porter SB, Sande M. Toxoplasmosis of the central nervous and the diagnostic value of polymerase chain reaction and
thalmol (Copenh). 1988;67:164-168. system in the acquired immunodeficiency syndrome. N Goldmann-Witmer coefficient in aqueous analysis. Am J
334. Foudrinier F, Villena I, Jaussaud R, et al. Clinical value of Engl J Med. 1992;327:1643-1648. Ophthalmol. 2007;144:781-785.
specific immunoglobulin E detection by enzyme-linked 357. Haverkos HW. Assessment of therapy for toxoplasma 382. Garweg JG, de Groot-Mijnes JD, Montoya JG. Diagnostic
immunosorbent assay in cases of acquired and congenital encephalitis. The TE Study Group. Am J Med. 1987;82: approach to ocular toxoplasmosis. Ocul Immunol Inflamm.
toxoplasmosis. J Clin Microbiol. 2003;41:1681-1686. 907-914. 2011;19:255-261.
335. Grover CM, Thulliez P, Remington JS, Boothroyd JD. Rapid 358. Levy RM, Rosenbloom S, Perrett LV. Neuroradiologic find- 383. Bretagne S, Costa JM, Keuntz M, et al. Late toxoplasmosis
prenatal diagnosis of congenital Toxoplasma infection by ings in AIDS: a review of 200 cases. Am J Neuroradiol. evidenced by PCR in marrow transplant reipient. Bone
using polymerase chain reaction and amniotic fluid. J Clin 1986;147:977-983. Marrow Transplant. 1995;15:809-811.
Microbiol. 1990;28:2297-2301. 359. Post MJ, Kursunoglu SJ, Hensley GT, et al. Cranial CT in 384. Turunen HJ, Leinikki PO, Saari KM. Demonstration of
336. Cinque P, Scarpellini P, Vago L, et al. Diagnosis of central acquired immunodeficiency syndrome: spectrum of dis- intraocular synthesis of immunoglobulin G Toxoplasma
nervous system complications in HIV-infected patients: eases and optimal contrast enhancement technique. AJR antibodies for specific diagnosis of toxoplasmic chorioreti-
cerebrospinal fluid analysis by the polymerase chain reac- Am J Roengenol. 1985;145:929-940. nitis by enzyme immunoassay. J Clin Microbiol. 1983;17:
tion. AIDS. 1997;11:1-17. 360. Levy RM, Mills CM, Posin JP, et al. The efficacy and clinical 988-992.
337. Dupouy-Camet J, Lavareda de Souza L, Maslo C, et al. impact of brain imaging in neurologically symptomatic 385. Rothova A, de Boer JH, Ten Dam-van Loon NH, et al.
Detection of Toxoplasma gondii in venous blood from AIDS patients: a prospective CT/MRI study. J Acquir Usefulness of aqueous humor analysis for the diagnosis of
AIDS patients by polymerase chain reaction. J Clin Micro- Immune Defic Syndr. 1990;3:461-471. posterior uveitis. Ophthalmology. 2008;115:306-311.
biol. 1993;31:1866-1869. 361. Post MJ, Chan JC, Hensley GT, et al. Toxoplasma encepha- 386. Montoya JG, Huffman HB, Remington JS. Use of the
338. Fricker-Hidalgo H, Brenier-Pinchart MP, Schaal JP, et al. litis in Haitian adults with acquired immunodeficiency VIDAS Toxo IgG Avidity test for the diagnosis of toxoplas-
Value of Toxoplasma gondii detection in one hundred syndrome: a clinical-pathologic-CT correlation. AJR Am J mic lymphadenopathy. Presented at 40th Annual Meeting
thirty-three placentas for the diagnosis of congenital toxo- Roengenol. 1983;140:861-868. of the Infectious Diseases Society of America (IDSA).
plasmosis. Pediatr Infect Dis J. 2007;26:845-846. 362. Ciricillo SF, Rosenblum ML. Use of CT and MR imaging Chicago, October 24-27, 2002.
339. Robert-Gangneux F, Dupretz P, Yvenou C, et al. Clinical to distinguish intracranial lesions and to define the 387. Wallon M, Franck J, Thulliez P, et al. Accuracy of real-time
relevance of placenta examination for the diagnosis of need for biopsy in AIDS patients. J Neurosurg. 1990;73: polymerase chain reaction for Toxoplasma gondii in amni-
congenital toxoplasmosis. Pediatr Infect Dis J. 2010;29: 720-724. otic fluid. Obstet Gynecol. 2010;115:727-733.
33-38. 363. Holloway RG, Mushlin AI. Intracranial mass lesions in 388. Sterkers Y, Pratlong F, Albaba S, et al. Novel interpretation
340. Sterkers Y, Varlet-Marie E, Marty P, Bastien P. Diversity and acquired immunodeficiency syndrome: using decision of molecular diagnosis of congenital toxoplasmosis accord-
evolution of methods and practices for the molecular diag- analysis to determine the effectiveness of stereotactic brain ing to gestational age at the time of maternal infection.
nosis of congenital toxoplasmosis in France: a 4-year biopsy. Neurology. 1996;46:1010-1015. J Clin Microbiol. 2012;50:3944-3951.
survey. Clin Microbiol Infect. 2010;16:1594-1602. 364. De La Paz R, Enzmann D. Neuroradiology of acquired 389. Remington JS, Araujo FG, Desmonts G. Recognition of
341. Torres E, Rivera R, Cardona N, et al. Evaluation of IgG immunodeficiency syndrome. In: Rosenblum ML, ed. different Toxoplasma antigens by IgM and IgG antibodies
anti-Toxoplasma avidity and PCR in the postnatal diagno- AIDS and the Nervous System. New York: Raven Press; in mothers and their congenitally infected newborns.
sis of congenital toxoplasmosis. Pediatr Infect Dis J. 2013; 1988:121-154. J Infect Dis. 1985;152:1020-1024.
32:693-695. 365. Luft BJ, Hafner R, Korzun AH, et al. Toxoplasmic encepha- 390. Rilling V, Dietz K, Krczal D, et al. Evaluation of a com-
342. Dannemann BR, Israelski DM, Leoung GS, et al. Toxo- litis in patients with the acquired immunodeficiency syn- mercial IgG/IgM Western blot assay for early postnatal
plasma serology, parasitemia and antigenemia in patients drome. N Engl J Med. 1993;329:995-1000. diagnosis of congenital toxoplasmosis. Eur J Clin Microbiol
at risk for toxoplasmic encephalitis. AIDS. 1991;5:1363- 366. Harris TM, Smith RR, Bognanno JR, Edwards MK. Infect Dis. 2003;22:174-180.
1365. Toxoplasmic myelitis in AIDS: gadolinium-enhanced MR. 391. Gallego-Marin C, Henao AC, Gomez-Marin JE. Clinical
343. Pelloux H, Dupouy-Camet J, Derouin F, et al, Group B-TS. J Comput Assist Tomogr. 1990;14:809-811. validation of a Western blot assay for congenital toxoplas-
A multicentre prospective study for the polymerase chain 367. O’Doherty MJ, Barrington SF, Campbell M, et al. PET scan- mosis and newborn screening in a hospital in Armenia
reaction detection of Toxoplasma gondii DNA in blood ning and the human immunodeficiency virus-positive (Quindio) Colombia. J Trop Pediatr. 2006;52:107-112.
samples from 186 AIDS patients with suspected toxoplas- patient. J Nucl Med. 1997;38:1575-1583. 392. Tridapalli E, Capretti M, Farneti G, et al. Congenital toxo-
mic encephalitis. AIDS. 1997;11:1888-1890. 368. Naddaf SY, Akisik MF, Aziz M, et al. Comparison between plasmosis: the importance of the Western blot method to
201
344. Patrat-Delon S, Gangneux JP, Lavoue S, et al. Correlation Tl-chloride and 99Tc(m)-sestamibi SPET brain imaging avoid unnecessary therapy in potentially infected new-
of parasite load determined by quantitative PCR to clinical for differentiating intracranial lymphoma from non- borns. Acta Paediatr. 2008;97:1298-1300.
outcome in a heart transplant patient with disseminated malignant lesions in AIDS patients. Nucl Med Commun. 393. L’Ollivier C, Wallon M, Faucher B, et al. Comparison of
toxoplasmosis. J Clin Microbiol. 2010;48:2541-2545. 1998;19:47-53. mother and child antibodies that target high-molecular-
345. Silveira C, Vallochi AL, Rodrigues da Silva U, et al. Toxo- 369. Ramsey RG, Gean AD. Central nervous system toxoplas- mass Toxoplasma gondii antigens by immunoblotting
plasma gondii in the peripheral blood of patients with mosis. Neuroimag Clin N Am. 1997;7:171-186. improves neonatal diagnosis of congenital toxoplasmosis.
acute and chronic toxoplasmosis. Br J Ophthalmol. 2011; 370. Rosenfeld SS, Hoffman JM, Coleman RE, et al. Studies of Clin Vaccine Immunol. 2012;19:1326-1328.
95:396-400. primary central nervous system lymphoma with [18F]- 394. Cazenave J, Forestier F, Bessieres M, et al. Contribution of
346. Bastien P. Molecular diagnosis of toxoplasmosis. Trans R fluorodeoxyglucose (FDG) PET. J Nucl Med. 1992;33:532- a new PCR assay to the prenatal diagnosis of congenital
Soc Trop Med Hyg. 2002;96(suppl 1):S205-S215. 536. toxoplasmosis. Prenat Diagn. 1992;12:119-127.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3153.e6
395. Fuentes I, Rodriguez M, Domingo CJ, et al. Urine sample Toxoplasma encephalitis in patients with AIDS. Antimicrob 439. de Gans J, Portegies P, Reiss P, et al. Pyrimethamine alone
used for congenital toxoplasmosis diagnosis by PCR. J Clin Agents Chemother. 1991;35:2049-2052. as maintenance therapy for central nervous system toxo-
Microbiol. 1996;34:2368-2371. 419. Chaisson RE, Benson CA, Dube MP, et al. Clarithromy- plasmosis in 38 patients with AIDS. J Acquir Immune Defic
396. van de Ven E, Melchers W, Galama J, et al. Identification cin therapy for bacteremic Mycobacterium avium complex Syndr. 1992;5:137-142.
Part III  Infectious Diseases and Their Etiologic Agents

of Toxoplasma gondii infections by BI gene amplification. disease. A randomized, double-blind, dose-ranging study 440. Katlama C, Mouthon B, Gourdon D, et al. Atovaquone as
J Clin Microbiol. 1991;19:2120-2124. in patients with AIDS. AIDS Clinical Trials Group Pro- long-term suppressive therapy for toxoplasmic encephalitis
397. Sterkers Y, Ribot J, Albaba S, et al. Diagnosis of congenital tocol 157 Study Team. Ann Intern Med. 1994;121:905- in patients with AIDS and multiple drug intolerance. AIDS.
toxoplasmosis by polymerase chain reaction on neonatal 911. 1996;10:1107-1112.
peripheral blood. Diagn Microbiol Infect Dis. 2011;71:174- 420. Cohn DL, Fisher EJ, Peng GT, et al. A prospective random- 441. Zolopa A, Andersen J, Powderly W, et al. Early antiretrovi-
176. ized trial of four three-drug regimens in the treatment ral therapy reduces AIDS progression/death in individuals
398. Guerina N, Hsu H-W, Meissner H, et al. Neonatal serologic of disseminated Mycobacterium avium complex disease with acute opportunistic infections: a multicenter random-
screening and early treatment for congenital Toxoplasma in AIDS patients: excess mortality associated with high- ized strategy trial. PloS One. 2009;4:e5575.
gondii infection. N Engl J Med. 1994;330:1858-1863. dose clarithromycin. Terry Beirn Community Programs 442. Carr A, Tindall B, Brew BJ, et al. Low-dose trimethoprim-
399. Jara M, Hsu HW, Eaton RB, Demaria A Jr. Epidemiology for Clinical Research on AIDS. Clin Infect Dis. 1999;29: sulfamethoxazole prophylaxis for toxoplasmic encephalitis
of congenital toxoplasmosis identified by population-based 125-133. in patients with AIDS. Ann Intern Med. 1992;117:106-111.
newborn screening in Massachusetts. Pediatr Infect Dis J. 421. Ward DJ. Dapsone/pyrimethamine for the treatment of 443. Bozzette S, Finkelstein D, Spector S, et al. A randomized
2001;20:1132-1135. Toxoplasmic encephalitis. Program and Abstracts of 8th trial of three antipneumocystis agents in patients with
400. Cohn J, McMeeking A, Cohen W, et al. Evaluation of the International Conference on AIDS. Amsterdam, July advanced human immunodeficiency virus infection. N
policy of empiric treatment of suspected Toxoplasma 19-24, 1992:B133. Engl J Med. 1995;332:693-699.
encephalitis in patients with the acquired immunodefi- 422. Pope-Pegram L, Gathe J Jr, Bohn B, et al. Treatment of 444. Podzamczer D, Salazar A, Jiménez J, et al. Intermittent
ciency syndrome. Am J Med. 1989;86:521-527. presumed central nervous system toxoplasmosis with trimethoprim-sulfamethoxazole compared with dapsone-
401. Carbone LG, Bendixen B, Appel GB. Sulfadiazine- doxycycline. Program and Abstracts of 7th International pyrimethamine for the simultaneous primary prophylaxis
associated obstructive nephropathy occurring in a patient Conference on AIDS. Florence, Italy, June 16-21, 1991: of Pneumocystis pneumonia and toxoplasmosis in patients
with the acquired immunodeficiency syndrome. Am J 188. infected with HIV. Ann Intern Med. 1995;122:755-761.
Kidney Dis. 1988;12:72-75. 423. Turett G, Pierone G, Masci J, Nicholas P. Failure of doxy- 445. Girard P-M, Landman R, Gaudebout C, et al. Dapsone-
402. Reboli AC, Mandler HD. Encephalopathy and psychoses cycline in the treatment of cerebral toxoplasmosis. Program pyrimethamine compared with aerosolized pentamidine as
associated with sulfadiazine in two patients with AIDS and and Abstracts of 6th International Conference on AIDS. a primary prophylaxis against Pneumocystis carinii pneu-
CNS toxoplasmosis. Clin Infect Dis. 1992;15:556-557. San Francisco, June 20-24, 1990:Th.B.479. monia and toxoplasmosis in HIV infection. N Engl J Med.
403. Leoung GS, Stanford JF, Giordano MF, et al. Trimethoprim- 424. Saba J, Morlat P, Raffi F, et al. Pyrimethamine plus azithro- 1993;328:1514-1520.
sulfamethoxazole (TMP-SMZ) dose escalation versus mycin for treatment of acute toxoplasmic encephalitis in 446. Schurmann D, Bergmann F, Albrecht H, et al. Effectiveness
direct rechallenge for Pneumocystis carinii pneumonia patients with AIDS. Eur J Clin Microbiol Infect Dis. 1993; of twice-weekly pyrimethamine-sulfadoxine as primary
prophylaxis in human immunodeficiency virus-infected 12:853-856. prophylaxis of Pneumocystis carinii pneumonia and toxo-
patients with previous adverse reaction to TMP-SMZ. 425. Jacobson JM, Hafner R, Remington J, et al. Dose-escalation, plasmic encephalitis in patients with advanced HIV infec-
J Infect Dis. 2001;184:992-997. phase I/II study of azithromycin and pyrimethamine for tion. Eur J Clin Microbiol Infect Dis. 2002;21:353-361.
404. Soffritti S, Ricci G, Prete A, et al. Successful desensitization the treatment of toxoplasmic encephalitis in AIDS. AIDS. 447. Zeller V, Truffot C, Agher R, et al. Discontinuation of sec-
to trimethoprim-sulfamethoxazole after allogeneic haema- 2001;15:583-589. ondary prophylaxis against disseminated Mycobacterium
topoietic stem cell transplantation: preliminary observa- 426. Pedrol E, Gonzales-Clemente JM, Gatell JM, et al. Central avium complex infection and toxoplasmic encephalitis.
tions. Med Pediatr Oncol. 2003;40:271-272. nervous system toxoplasmosis in AIDS patients: efficacy Clin Infect Dis. 2002;34:662-667.
405. Coppola S, Angarano G, Monno L, et al. Adverse effects of of an intermittent maintenance therapy. AIDS. 1990;4: 448. Mussini C, Pezzotti P, Govoni A, et al. Discontinuation of
clindamycin in the treatment of cerebral toxoplasmosis in 511-517. primary prophylaxis for Pneumocystis carinii pneumonia
AIDS patients. Program and Abstracts of 7th International 427. Podzamczer D, Miró J, Bolao F, et al. Twice-weekly main- and toxoplasmic encephalitis in human immunodeficiency
Conference on AIDS. Florence, Italy, June 16-21, 1991:265. tenance therapy with sulfadiazine-pyrimethamine to virus type I-infected patients: the changes in opportunistic
406. Torre D, Casari S, Speranza F, et al. Randomized trial of prevent recurrent toxoplasmic encephalitis in patients with prophylaxis study. J Infect Dis. 2000;181:1635-1642.
trimethoprim-sulfamethoxazole versus pyrimethamine- AIDS. Ann Intern Med. 1995;123:175-180. 449. Baden LR, Katz JT, Franck L, et al. Successful toxoplasmo-
sulfadiazine for therapy of toxoplasmic encephalitis in 428. Podzamczer D, Miro JM, Ferrer E, et al. Thrice-weekly sis prophylaxis after orthotopic cardiac transplantation
patients with AIDS. Italian Collaborative Study Group. sulfadiazine-pyrimethamine for maintenance therapy of with trimethoprim-sulfamethoxazole. Transplantation.
Antimicrob Agents Chemother. 1998;42:1346-1349. toxoplasmic encephalitis in HIV-infected patients. Spanish 2003;75:339-343.
407. Alavi SM, Alavi L. Treatment of toxoplasmic lymphadenitis Toxoplasmosis Study Group. Eur J Clin Microbiol Infect Dis. 450. Durant J, Hazime F, Carles M, et al. Prevention of Pneumo-
with co-trimoxazole: double-blind, randomized clinical 2000;19:89-95. cystis carinii pneumonia and of cerebral toxoplasmosis by
trial. Int J Infect Dis. 2010;14(suppl 3):e67-e69. 429. Heald A, Flepp M, Chave J-P, et al. Treatment for cerebral roxithromycin in HIV-infected patients. Infection. 1995;23:
408. Carey RM, Kimball AC, Armstrong D, Lieberman PH. toxoplasmosis protects against Pneumocystis carinii pneu- S33-S38.
Toxoplasmosis. Clinical experiences in a cancer hospital. monia in patients with AIDS. Ann Intern Med. 1991;115: 451. Jacobson M, Besch C, Child C, et al. Primary prophylaxis
Am J Med. 1973;54:30-38. 760-763. with pyrimethamine for toxoplasmic encephalitis in
409. Katlama C, De Wit S, O’Doherty E, et al. Pyrimethamine- 430. Duval X, Pajot O, Le Moing V, et al. Maintenance therapy patients with advanced human immunodeficiency virus
clindamycin vs. pyrimethamine-sulfadiazine as acute and with cotrimoxazole for toxoplasmic encephalitis in the disease: results of a randomized trial. J Infect Dis. 1994;
long-term therapy for toxoplasmic encephalitis in patients era of highly active antiretroviral therapy. AIDS. 2004;18: 169:384-394.
with AIDS. Clin Infect Dis. 1996;22:268-275. 1342-1344. 452. Klinker H, Langmann P, Richter E. Pyrimethamine alone
410. Luft BJ, Remington JS. AIDS commentary. Toxoplasmic 431. Remington JS, Vilde JL, Antunes F, et al. Clindamycin for as prophylaxis for cerebral toxoplasmosis in patients with
encephalitis. J Infect Dis. 1988;157:1-6. toxoplasmosis encephalitis in AIDS. Lancet. 1991;338: advanced HIV infection. Infection. 1996;4:324-328.
411. Dannemann BR, McCutchan JA, Israelski DA, et al. Treat- 1142-1143. 453. Raffi F, Struillou L, Ninin E, et al. Breakthrough cerebral
ment of toxoplasmic encephalitis in patients with AIDS: a 432. Kaplan JE, Benson C, Holmes KH, et al. Guidelines for toxoplasmosis in patients with AIDS who are being treated
randomized trial comparing pyrimethamine plus clinda- prevention and treatment of opportunistic infections in with clarithromycin. Clin Infect Dis. 1995;20:1076-1077.
mycin to pyrimethamine plus sulfadiazine. Ann Intern HIV-infected adults and adolescents: recommendations 454. Leport C, Vilde JL, Katlama C, et al. Failure of spiramycin
Med. 1992;116:33-43. from CDC, the National Institutes of Health, and the HIV to prevent neurotoxoplasmosis in immunosuppressed
412. Canessa A, Del Bono V, De Leo P, et al. Cotrimoxazole Medicine Association of the Infectious Diseases Society of patients. Med Clin North Am. 1986;70:677-692.
therapy of Toxoplasma gondii encephalitis in AIDS America. MMWR Recomm Rep. 2009;58:1-207; quiz 455. Jacobson M, Besch C, Child C, et al. Toxicity of clindamy-
patients. Eur J Clin Microbiol Infect Dis. 1992;11:125-130. CE1-4. cin as prophylaxis for AIDS-associated toxoplasmic
413. Solbreux P, Sonnet J, Zech F. A retrospective study about 433. Köppen S, Grunewald T, Jautzke G, et al. Prevention of encephalitis. Lancet. 1992;339:333-334.
the use of cotrimoxazole as diagnositc support and treat- Pneumocystis carinii pneumonia and toxoplasmic encepha- 456. Cochereau-Massin I, LeHoang P, Lautier-Frau M, et al.
ment of suspected cerebral toxoplasmosis in AIDS. Acta litis in human immunodeficiency virus infected patients: Ocular toxoplasmosis in human immunodeficiency virus-
Clin Belg. 1990;45:85-96. a clinical approach comparing aerosolized pentamidine infected patients. Am J Ophthalmol. 1992;114:130-135.
414. Torre D, Speranza F, Martegani R, et al. A retrospective and pyrimethamine/sulfadoxine. Clin Investig. 1992;70: 457. Friedman AH, Orellana J, Gagliuso DJ, Teich SA. Ocular
study of treatment of cerebral toxoplasmosis in AIDS 508-512. toxoplasmosis in AIDS patients. Trans Am Ophthalmol Soc.
patients with trimethoprim-sulphamethoxazole. J Infect. 434. Clotet B, Sirera G, Romeu J, et al. Twice-weekly dapsone- 1990;88:63-88.
1998;37:15-18. pyrimethamine for preventing PCP and cerebral toxoplas- 458. Schnapp L, Geaghan S, Campagna A, et al. Toxoplasma
415. Kovacs JA. Efficacy of atovaquone in treatment of toxoplas- mosis. AIDS. 1991;5:601-602. gondii pneumonitis in patients infected with the human
mosis in patients with AIDS. Lancet. 1992;340:637-638. 435. Opravil M, Hirschel B, Lazzarin A, et al. Once-weekly immunodeficiency virus. Arch Intern Med. 1992;152:1073-
416. Torres R, Weinberg W, Stansell J, et al. Atovaquone for administration of dapsone/pyrimethamine vs. aerosolized 1076.
salvage treatment and suppression of toxoplasmic encepha- pentamidine as a combined prophylaxis for Pneumocystis 459. Kim SJ, Scott IU, Brown GC, et al. Interventions for
litis in patients with AIDS. Clin Infect Dis. 1997;24:422- carinii pneumonia and toxoplasmic encephalitis in human Toxoplasma retinochoroiditis: a report by the American
429. immunodeficiency virus-infected patients. Clin Infect Dis. Academy of Ophthalmology. Ophthalmology. 2013;120:
417. Chirgwin K, Hafner R, Leport C, et al. Randomized phase 1995;20:531-541. 371-378.
II trial of atovaquone with pyrimethamine or sulfadiazine 436. Torres R, Barr M, Thorn M, et al. Randomized trial of 460. Holland GN, Lewis KG. An update on current practices in
for treatment of toxoplasmic encephalitis in patients with dapsone and aerosolized pentamidine for the prophylaxis the management of ocular toxoplasmosis. Am J Ophthal-
acquired immunodeficiency syndrome: ACTG 237/ANRS of Pneumocystis carinii pneumonia and toxoplasmic mol. 2002;134:102-114.
039 Study. AIDS Clinical Trials Group 237/Agence Natio- encephalitis. Am J Med. 1993;95:573-583. 461. Holland GN. Ocular toxoplasmosis: a global reassessment,
nale de Recherche sur le SIDA, Essai 039. Clin Infect Dis. 437. Navin TR, Miller KD, Satriale RF, Lobel HO. Adverse reac- Part I. Epidemiology and course of disease. Am J Ophthal-
2002;34:1243-1250. tions associated with pyrimethamine-sulfadoxine prophy- mol. 2003;136:973-988.
417a.  Calderón MM, Penzak SR, Pau AK, et al. Efavirenz but laxis for Pneumocystis carinii infections in AIDS. Lancet. 462. Holland GN. Ocular toxoplasmosis: a global reassessment,
not atazanavir/ritonavir significantly reduces atovaquone 1985;1:1332. Part II. Disease manifestations and management. Am J
concentrations in HIV-infected subjects. Clin Infect Dis. 438. Maslo C, Matheron S, Saimot AG. Cerebral toxoplasmosis: Ophthalmol. 2004;137:1-17.
2016;62:1036-1042. assessment of maintenance therapy. Program and Abstracts 463. Soheilian M, Sadoughi MM, Ghajarnia M, et al. Prospec-
418. Fernandez-Martin J, Leport C, Morlat P, et al. of 8th International Conference on AIDS. Amsterdam, July tive randomized trial of trimethoprim/sulfamethoxazole
Pyrimethamine-clarithromycin for therapy of acute 19-24, 1992:B123. versus pyrimethamine and sulfadiazine in the treatment

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
3153.e7
of ocular toxoplasmosis. Ophthalmology. 2005;112:1876- 469. Forestier F, Daffos F, Rainaut M, et al. Suivi therapeutique transmission rate and improves clinical outcome at age 3
1882. foetomaternel de la spiramycine en cours de grossesse. years. Clin Infect Dis. 2013;56:1223-1231.
464. Baharivand N, Mahdavifard A, Fouladi RF. Intravitreal Arch Fr Pediatr. 1987;44:539-544. 477. Garweg JG, Scherrer J, Wallon M, et al. Reactivation of
clindamycin plus dexamethasone versus classic oral 470. Gilbert R, Gras L; European Multicentre Study on Con- ocular toxoplasmosis during pregnancy. BJOG. 2005;112:

Chapter 280  Toxoplasma gondii


therapy in toxoplasmic retinochoroiditis: a prospective genital Toxoplasmosis. Effect of timing and type of treat- 241-242.
randomized clinical trial. Int Ophthalmol. 2013;33:39-46. ment on the risk of mother to child transmission of 478. McAuley J, Boyer KM, Patel D, et al. Early and longitudinal
465. Soheilian M, Ramezani A, Azimzadeh A, et al. Random- Toxoplasma gondii. BJOG. 2003;110:112-120. evaluations of treated infants and children and untreated
ized trial of intravitreal clindamycin and dexamethasone 471. Gilbert RE, Gras L, Wallon M, et al. Effect of prenatal treat- historical patients with congenital toxoplasmosis: the
versus pyrimethamine, sulfadiazine, and prednisolone in ment on mother to child transmission of Toxoplasma Chicago Collaborative Treatment Trial. Clin Infect Dis.
treatment of ocular toxoplasmosis. Ophthalmology. 2011; gondii: retrospective cohort study of 554 mother-child 1994;18:38-72.
118:134-141. pairs in Lyon, France. Int J Epidemiol. 2001;30:1303- 479. McGee T, Wolters C, Stein L, et al. Absence of sensori-
466. Pearson PA, Piracha AR, Sen HA, Jaffe GJ. Atovaquone 1308. neural hearing loss in treated infants and children with
for the treatment of Toxoplasma retinochoroiditis in 472. Gilbert R, Dunn D, Wallon M, et al. Ecological comparison congenital toxoplasmosis. Otolarygol Head Neck Surg.
immunocompetent patients. Ophthalmology. 1999;106: of the risks of mother-to-child transmission and clinical 1992;106:75-80.
148-153. manifestations of congenital toxoplasmosis according to 480. Mets MB, Holfels E, Boyer KM, et al. Eye manifestations of
467. Bosch-Driessen LH, Verbraak FD, Suttorp-Schulten MS, prenatal treatment protocol. Epidemiol Infect. 2001;127: congenital toxoplasmosis. Am J Opthalmol. 1996;122:309-
et al. A prospective, randomized trial of pyrimethamine 113-120. 324.
and azithromycin vs pyrimethamine and sulfadiazine for 473. Foulon W, Villena I, Stray-Pedersen B, et al. Treatment of 481. Roizen N, Swisher CN, Stein MA, et al. Neurologic and
the treatment of ocular toxoplasmosis. Am J Ophthalmol. toxoplasmosis during pregnancy: a multicenter study of developmental outcome in treated congental toxoplasmo-
2002;134:34-40. impact on fetal transmission and children’s sequelae at age sis. Pediatrics. 1995;95:11-20.
467a.  Silveira C, Muccioli C, Nussenblatt R, et al. The effect of 1 year. Am J Obstet Gynecol. 1999;180:410-415. 482. Wreghitt TG, Hakim M, Gray JJ, et al. Toxoplasmosis in
long-term intermittent trimethoprim/sulfamethoxazole 474. Thulliez P. Commentary: efficacy of prenatal treatment for heart and heart lung transplant recipients. J Clin Pathol.
treatment on recurrences of toxoplasmic retinochoroiditis: toxoplasmosis: a possibility that cannot be ruled out. Int J 1989;42:194-199.
10 years of follow-up. Ocul Immunol Inflamm. 2015;23: Epidemiol. 2001;30:1315-1316. 483. Daffos F, Forestier F, Capella-Pavlovsky M, et al. Prenatal
246-247. 475. Moncada PA, Montoya JG. Toxoplasmosis in the fetus and management of 746 pregnancies at risk for congenital toxo-
468. Valentini P, Annunziata ML, Angelone DF, et al. Role of newborn: an update on prevalence, diagnosis and treat- plasmosis. N Engl J Med. 1988;318:271-275.
spiramycin/cotrimoxazole association in the mother-to- ment. Expert Rev Anti Infect Ther. 2012;10:815-828. 484. Desmonts G, Daffos F, Forestier F, et al. Prenatal diag-
child transmission of toxoplasmosis infection in preg- 476. Wallon M, Peyron F, Cornu C, et al. Congenital Toxo- nosis of congenital toxoplasmosis. Lancet. 1985;1:500-
nancy. Eur J Clin Microbiol Infect Dis. 2009;28:297-300. plasma infection: monthly prenatal screening decreases 504.

Downloaded for akses clinicalkey akses (aksesclinicalkey@gmail.com) at Universitas Warmadewa from ClinicalKey.com by Elsevier on October 11, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.

Anda mungkin juga menyukai