Вы находитесь на странице: 1из 15

Reproductive Toxicology 21 (2006) 458–472

Review

Congenital toxoplasmosis—prenatal aspects of Toxoplasma gondii infection


Efrat Rorman a,∗ , Chen Stein Zamir b , Irena Rilkis a,c , Hilla Ben-David a
a National Public Health Laboratory, Ministry of Health, P.O. Box 8255, Tel Aviv 61082, Israel
b District Health Office, Ministry of Health, Jerusalem, Israel
c National Toxoplasmosis Reference Center, Ministry of Health, Israel

Received 26 August 2004; received in revised form 11 October 2005; accepted 24 October 2005
Available online 28 November 2005

Abstract
Toxoplasma gondii (T. gondii) is the cause of toxoplasmosis. Primary infection in an immunocompetent person is usually asymptomatic.
Serological surveys demonstrate that world-wide exposure to T. gondii is high (30% in US and 50–80% in Europe). Vertical transmission from a
recently infected pregnant woman to her fetus may lead to congenital toxoplasmosis. The risk of such transmission increases as primary maternal
infection occurs later in pregnancy. However, consequences for the fetus are more severe with transmission closer to conception. The timing of
maternal primary infection is, therefore, critically linked to the clinical manifestations of the infection. Fetal infection may result in natural abortion.
Often, no apparent symptoms are observed at birth and complications develop only later in life. The laboratory methods of assessing fetal risk of
T. gondii infection are serology and direct tests.
Screening programs for women at childbearing age or of the newborn, as well as education of the public regarding infection prevention, proved
to be cost-effective and reduce the rate of infection.
The impact of antiparasytic therapy on vertical transmission from mother to fetus is still controversial. However, specific therapy is recommended
to be initiated as soon as infection is diagnosed.
© 2005 Elsevier Inc. All rights reserved.

Keywords: Toxoplasmosis; Toxoplasma gondii; Congenital infection; Diagnosis; Treatment; Epidemiology

Contents

1. Case report . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 459


2. The parasite . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 459
2.1. Life cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 459
2.2. Mechanism of infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 460
2.3. Virulence of T. gondii strains . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 460
3. Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 460
4. Congenital toxoplasmosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 461
4.1. Incidence and prevalence in pregnant women and infants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 461
4.2. Diagnostic evaluation, manifestation and consequences . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 461
4.3. Prenatal laboratory diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463
4.3.1. Sabin Feldman dye test (SFDT) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463
4.3.2. Enzyme immunoassays (EIA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465
4.3.3. Immunosorbent agglutination assay test (IAAT) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465
4.3.4. Indirect fluorescent assay (IFA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465
4.3.5. Avidity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 465
4.3.6. Animal and cell culture inoculation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 466

∗ Corresponding author. Tel.: +972 50 6242904; fax: +972 3 6826996.


E-mail address: efrat.rorman@phlta.health.gov.il (E. Rorman).

0890-6238/$ – see front matter © 2005 Elsevier Inc. All rights reserved.
doi:10.1016/j.reprotox.2005.10.006
E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472 459

4.3.7. Molecular diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 466


4.4. Laboratory diagnosis of infants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 467
4.4.1. Western blots . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 467
5. Treatment of congenital toxoplasmosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 467
6. Prevention . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 468
6.1. Primary prevention . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 468
6.1.1. Vaccine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 468
6.2. Secondary prevention – screening . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 468
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 468
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 469

1. Case report normal. Cranial ultrasonography, brain stem evoked response


(BERA), audiometry and eye examination were all normal. Tests
A 26-year-old woman from a rural village in northern Israel for T. gondii in the infant included: PCR of CSF—negative,
presented with cervical lymphadenopathy during the 13th week immuno-sorbent agglutination assays (IgM-ISAGA)—negative
of her first pregnancy. The woman was otherwise healthy and and Sabin Feldman Dye Test (SFDT)—positive (250 IU/ml),
without any symptoms. She was followed up by her primary probably reflecting maternal antibodies transfer. Despite the
care physician and as the lymphadenopathy did not resolve, serological indicators of maternal infection (most probably
was sent for surgical consultation during the 26th week of towards the end of the first trimester) and positive PCR of the
pregnancy. The surgeon referred her to laboratory tests for Tox- amniotic fluid, there was no evidence of congenital toxoplasmo-
oplasma gondii-specific antibodies and for various other infec- sis in the neonate. The infant was treated with the same thera-
tions. The results obtained from testing a serum sample from peutic protocol as the mother planned to be continued until the
the 26th week of gestation, performed at the Israel National age of 1 year. Medical evaluation, auditory and ophthalmic tests
Toxoplasmosis Reference Center were: positive for total T. at the age of 4 and 8 months revealed normal physical growth
gondii-specific immunoglobulins (Ig) (250 IU/ml by Sabin Feld- and development and intensive follow-up continues (at the age
man Dye Test) and for T. gondii-specific IgM antibodies (by of 6 months laboratory analysis was reported to be normal).
ELFA, Enzyme-Linked Fluorescent immuno-Assay) with low This case demonstrates the complexity of establishing clin-
IgG avidity (0.027). These results were reported and an addi- ical diagnosis and interpretation of laboratory results in regard
tional serum sample, as well as an earlier sample (whether to T. gondii infection in pregnancy. The favourable outcome
available) were requested. Blood samples were subsequently despite the timing of infection may be attributed to providing anti
delivered to our laboratory; from the 12th (sample drawn as part parasitic therapy, although the specific role of therapy or other
of the routine pregnancy follow-up) and from the 34th weeks unknown variables is unclear. Since many T. gondii infections
of pregnancy. The results of the earlier sample were negative are sub-clinical or present with non-specific signs, physicians
for both total Ig and IgM antibodies. The results from the 34th should be able to integrate clinical and laboratory data in order
week were positive for total T. gondii-specific immunoglobu- to make diagnostic and therapeutic decisions.
lins (250 IU/ml by Sabin Feldman Dye Test) and negative for
T. gondii-specific IgM antibodies (by ELFA, Enzyme-Linked 2. The parasite
Fluorescent immuno-Assay) with low IgG avidity (0.055).
The sum interpretation of the three above tests results of T. gondii is a member of the phylum Apicomplexa, order Coc-
the 12th, 26th and 34th week of pregnancy was consistent cidia, which are all obligate intracellular protozoan parasites.
with definite recent T. gondii infection (seroconversion, constant Other members of this phylum include known human pathogens
high T. gondii-specific immunoglobulins, emergence and disap- such as Plasmodium (malaria) and Cryptosporidium.
pearance of IgM, low avidity and cervical lymphadenopathy).
Amniocentesis was performed during the 35th week of preg- 2.1. Life cycle
nancy and PCR result for T. gondii DNA in the amniotic fluid
was positive. The life cycle of T. gondii consists of two stages—asexual and
The woman was referred for follow-up at a high risk preg- sexual: the asexual stage takes place in the intermediate hosts,
nancy clinic in a tertiary medical center. Anti-T. gondii therapy which are mammals or birds. During this phase rapid intracellu-
including Pyrimethamine, Sulfadiazine and folinic acid was lar growth of the parasite as tachyzoite takes place (generation
started and continued until birth. The pregnancy course was time in vitro is 6–8 h). The oval or crescent-shaped tachyzoites
otherwise uneventful and fetal growth assessment through ultra- can infect and multiply in almost any nucleated mammalian or
sound follow-up did not reveal any abnormality. During the avian cell [1]. Following accumulation (64–128), tachyzoites are
38th week of pregnancy a female infant was born by sponta- secreted into the blood stream [2] and spread in the body, leading
neous delivery. Birth weight was 2830 g and head circumfer- to development of an acute disease (parasitemia). The normal
ence was 33 cm. Physical examination was normal. Laboratory immune response and transformation of the tachyzoite into cyst-
tests including complete blood count, glucose, electrolytes, liver forming bradyzoites limit the acute stage and establish a chronic
function tests and cerebro-spinal fluid (CSF) tests were all infection. Bradyzoites differ from tachyzoites mainly in their
460 E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472

extremely slow multiplication rate (their name reflects this slow genome of T. gondii, consisting of 14 chromosomes, is currently
process) and in the distinct set of proteins they express [1,3–5]. being investigated and sequenced [26] (http://ToxoDB.org).
The cysts are formed mainly in neural and muscular tissues,
especially brain, skeletal and cardiac muscles, and can persist, 2.3. Virulence of T. gondii strains
inactivated, in the body for a very long time. In the immunocom-
promised patient the release of bradyzoites from the cyst may Clinical manifestations and severity of illness following
cause acute encephalitis. infection are affected by features of the interaction between the
The sexual stage takes place in the intestine of the definitive parasite and the host and include strain virulence, inoculum size,
host. Known definitive hosts are members of the feline fam- route of infection, competence of the host’s immune response
ily, predominantly domestic cats. When bradyzoites or oocytes (both cellular and humoral), integrity of the host’s mucosal and
are ingested by a feline, formation of oocytes proceeds in the epithelial barriers, host’s age and genetic background [27]. Var-
epithelium of the small intestine. Several million unsporulated ious strains of T. gondii have long been known to differ in
oocytes may be released in the feces of a single cat over a period virulence and pathogenicity [28,29]. These strains can be classi-
3–18 days, depending on the stage of T. gondii ingested [1]. fied by immunologic assays, isoenzyme analysis and molecular
Under mild environmental conditions oocytes may sporulate analysis [30–33]. There are three T. gondii clonal lineages, of
within a 3-week period [6], then infecting humans and other them one carries conserved genetic loci, suspected of coding
intermediate hosts. Oocysts can spread in the environment and for the virulence trait [24]. Grigg et al. [34] demonstrated that a
contaminate water, soil, fruits, vegetables and herbivores fol- sexual recombination, performed in vitro, between the two rela-
lowing consumption of infected plant material. Investigation of tively avirulent strains can give rise to the virulent strain. This is
outbreaks of toxoplasmosis has led to recovery of oocytes from in accordance with polymorphism analysis of the three T. gondii
soil [7] but not from water [8–10]. Oocytes have been found to strains, which indicated that they emerged within the last 10,000
be very stable, especially in warm and humid environments, and years, following a single genetic cross [34,35]. Acquisition of
resistant to many disinfecting agents [11], but survive poorly in an efficient mechanism to spread by direct oral transmission,
arid, cold climates [12]. bypassing a sexual phase, leads to successful clonal expansion
of this virulent lineage [35,36].
2.2. Mechanism of infection Genetic background plays a significant role in increased sus-
ceptibility to T. gondi in humans; HLA-DQ3 appears to be
T. gondii has been shown to migrate over long distances in the a genetic marker associated with susceptibility to developing
host’s body; crossing biological barriers, actively enter the blood toxoplasma-dependent encephalitis [37,38].
stream, invade cells and cross substrates and non-permissive
biological sites such as the blood-brain-barrier, the placenta and 3. Epidemiology
the intestinal wall. At the same time, the parasite minimizes
exposure to the host’s immune response, by rapidly entering and T. gondii infection is most frequently caused by ingestion
exiting cells. These two functions share common mechanisms of row or undercooked meat, which carries tissue cysts, by
which depend on Ca2+ regulation [13]. consuming infected water or food or by accidental intake of
Unlike many bacteria and viruses, T. gondii actively enters contaminated soil. Toxoplasmosis is also an occupational haz-
the cell, in a mechanism which is mediated by the para- ard for laboratory workers. A total of 47 laboratory-acquired
sites’ cytoskeleton and regulated by a parasite-specific calcium- cases have been reported, 81% of them were symptomatic cases
depended secretion pathway [2,14]. The first step of cell invasion [39].
by T. gondii is recognition of an attachment point. The two Tender et al. [40] collected data of nation-wide T. gondii sero-
special organelles involved in this invasion process, rhoptries prevalence in women at child-bearing age (1990–2000). The
and micronemes, each discharging proteins during the process rates of positive sero-prevalence, were 58% in Central Euro-
[5,15]. Following the rapid cellular invasion the parasite resides pean countries, 51–72% in several Latin-American countries
within a vacuole, derived primarily from the host cell’s plasma and 54–77% in West African countries. Low seroprevalence,
membrane [2,16]. The active motion of T. gondii, called “glid- 4–39%, was reported in southwest Asia, China and Korea as
ing”, occurs with no major changes in cell shape. It is fast (about well as in cold climate areas such as Scandinavian countries
10 times faster than the “crawling” rate of amoeboid cells), and (11–28%). In the US 15% of females at childbearing age were
consists of both circular gliding in a counter-clockwise direction found to be seropositive [41]. It should be noted that seropositive
and clockwise helical gliding [17–21]. As an obligatory parasite, prevalence in the same country may differ among populations
it’s invasive capabilities play an important role in virulence and or geographical regions and world-wide prevalence is higher in
pathogenicity, since it can only survive intracellularly where it older populations.
gets nutrients and escapes from the host’s immune response [22]. In a limited case–control study that included six large Euro-
The most virulent T. gondii strain has been shown to exhibit supe- pean centers it was shown that the consumption of undercooked
rior migratory capacity [23] and a subpopulation of this strain meat was the major risk factor for toxoplasmosis infection [42].
displays a special, long distance migration phenotype [14]. The Another study aimed to determine the prevalence of T. gondii
ability to cross biological barriers is associated with acute vir- in edible meat tested 71 meat samples from commercial sources
ulence and is linked to genes on chromosome VII [24,25]. The in the UK for the parasite—positive results were found in 27
E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472 461

samples. Twenty-one of these contaminated meat samples car- disease, resulting in brain lesions or diffuse encephalitis. Other
ried the virulent T. gondii type I [43]. Although cats play a organs, such as the heart, lung, liver, and retina may also be
definite role in the epidemiology of toxoplasmosis, no signif- involved. Most of these cases result from reactivation of latent
icant correlation between human toxoplasmosis infection and infection [54] although re-infection with a different T. gondii
cat ownership could be proven [44]. Furthermore, the oocytes strain in the transplanted organs may also occur.
are not found on cat fur but rather are buried in the soil as they
are shed with cat faeces [45–47]. 4.1. Incidence and prevalence in pregnant women and
Data regarding seroprevalence of specific T. gondii anti- infants
bodies in the Israeli population are based on several regional
surveys performed in collaboration with the Israeli National The disease is caused by vertical transmission of T. gondii
Toxoplasmosis Reference Center. The prevalence in certain sub- from a seronegative pregnant woman, who is acutely infected
populations of pregnant women in northern Israel had been with T. gondii to her fetus.
reported to be 21% on average and the incidence rate of infection The prevalence of T. gondii and its incidence of human infec-
acquired during pregnancy estimated as 1.4% [48]. tion vary widely amongst various countries. Worldwide, 3–8
Human contact with infected oocyst from contaminated soil infants per 1000 live births are infected in utero [55]. Multiple
[7,49,50] and water [8–10] were associated with several reported factors are associated with the occurrence of congenital toxo-
epidemics caused by T. gondii. Only in one case were T. plasmosis infection, including route of transmission, climate,
gondii oocysts recovered from the soil—the suspected source cultural behaviour, eating habits and hygienic standards. This
of infection [7]. There are ongoing efforts to develop sensi- combination leads to marked differences even among developed
tive detection techniques for environmental samples [11,51,52]. nations. For example, the incidence of congenital infection in
Unfortunately, isolation of oocytes from such samples is dif- Belgium and France is 2–3 cases per 1000 live births—markedly
ficult, since infectious doses are small while large volume of higher than the US incidence, which is between 1 in 10,000 to
sample is required for isolation of the organism. In addition, 1 in 1000 live births [47,56].
there is a lag period between the time of infection and the time In a research conducted in Goiania, Brazil, a region with
that the contaminated source is tested, further reducing the like- a relatively high seroconversion rate, pregnant women were
lihood of recovery of oocytes from the suspected environment found to have a 2.2 times higher risk for seroconversion than
during epidemiological investigation. non-pregnant women of equivalent characteristics. In addition,
T. gondii was reported to cause 0.8% of the total food-borne amongst pregnant women, adolescents were shown to have the
illnesses attributed to a known pathogen, and 20.7% of the total highest risk for seroconversion [57]. The authors hypothesized
food-borne mortality caused by a known pathogen, in the United that higher vulnerability to T. gondii infection during pregnancy
States in 1996–1997. Many of these cases involved HIV-infected may be due to a combination of pregnancy associated immuno-
patients [53]. suppression as well as hormonal changes.
The largest reported toxoplasmosis outbreak resulting from Only a few cases of congenital toxoplasmosis transmitted
contaminated water occurred in British Columbia and caused by mothers who were infected prior to conception have actu-
acute infection in 100 people; 19 with retinitis and 51 with lym- ally been reported [58–60]. One such case published recently
phadenopathy. The likely source was a municipal water system involved a woman who had ocular toxoplasmosis 20 years prior
that used unfiltered, chloraminated surface water [10]. There was to giving birth to a newborn, who suffered from congenital tox-
also a seasonal correlation to rainfall and turbidity in this water oplasmosis. The mother had a “toxoplasmic scar” in the retina
reservoir. In another small outbreak North of Rio de Janeiro, and was tested positive for specific toxoplasma IgG antibodies.
Brazil, the source of the parasite was traced to an unfiltered water The newborn was found to be positive for both IgG and IgM
source. It was also linked to high prevalence of seropositivity in antibodies and had a macular scar on the retina, typical to tox-
this region of low socio-economic background [8]. oplasmosis, as well as a calcified brain granuloma. [59]. Such
a case could be attributed to re-infection with a different, more
4. Congenital toxoplasmosis virulent strain or by reactivation of a chronic disease[58].
Chronically infected women, who are immunodeficienct,
Most cases of acquired toxoplasma infection are asymp- may also transmit the infection to their fetus; the risk of this
tomatic and self-limited; hence many cases remain undiagnosed. occurrence is difficult to quantify, but it is probably low. Latent
The incubation period of acquired infection is estimated to be T. gondii infection may be reactivated in immunodeficient indi-
within a range of 4–21days (7 days on average) [10]. When viduals (such as HIV-infected women) and result in congenital
symptomatic infection does occur the only clinical findings may transmission of the parasite [61].
be focal lymphadenopathy, most often involving a single site
around the head and neck. Less commonly, acute infection is 4.2. Diagnostic evaluation, manifestation and
accompanied by a mononucleosis-like syndrome characterized consequences
by fever, malaise, sore throat, headache and an atypical lympho-
cytosis on peripheral blood smear [54]. In immunocompromised The diagnostic evaluation of T. gondii is part of routine preg-
patients, most commonly HIV infected and organ transplant nancy follow-up and differential diagnosis of intrauterine infec-
recipients, T. gondii may cause a severe central nervous system tion. Intrauterine ultrasonographic findings of T. gondii infection
462 E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472

are usually non-specific and in most cases no pathological evi- ties. They concluded that positive screening results must be care-
dences are revealed. In certain cases the ultrasonographic find- fully confirmed [66]. Laboratory methods and their implications
ings may include: intracranial calcifications, echogenic streaks, in supporting evidence-based diagnoses are discussed below.
microcephalus, ventricular dilatation and hydrocephalus [62]. The risk of fetal infection is multifactorial, depending on the
Gay-Andrieu et al. [63] described two cases of intrauterine time of maternal infection, immunological competence of the
infection in which the diagnosis was based upon hydrocephalus mother during parasitemia, parasite load and strain’s virulence
in fetal ultrasound, even though PCR of amniotic fluid was [40]. The probability of fetal infection is only 1% when pri-
negative in both cases. The authors emphasized that hydro- mary maternal infection occurs during the preconception period
cephalus is the most frequent lesion detected by fetal ultra- but increases as pregnancy progresses; infection acquired dur-
sound, reflecting the pathological process taking place within ing the first trimester by women not treated with anti-T. gondii
several months post-infection in cases of intrauterine infection drugs results in congenital infection in 10 to 25% of cases.
of T. gondii. Additional ultrasonographic findings may include For infections occurring during the second and third trimesters,
hepatomegaly, splenomegaly, ascitic fluid, cardiomegaly and the incidence of fetal infection ranges between 30–54% and
placental abnormalities [55,64]. Safadi et al. [65] followed 43 60–65%, respectively [54].
children with congenital toxoplasmosis for a period of at least The consequences are more severe when fetal infection
5 years. Most of them (88%) had sub-clinical presentation occurs in early stages of pregnancy, when it can cause miscar-
at birth. The most common neurological manifestation was a riage (natural abortion or death occurs in 10% of pregnancies
delay in neuro-psychomotor development. Half of the children infected with T. gondii [67]), severe disease, intra-uterine growth
developed neurological manifestations, 7 children had neuro- retardation or premature birth. A multi-centre prospective cohort
radiologic alterations in skull radiography, and 33 children in study evaluated the association between congenital toxoplas-
tomography. Notably, cerebral calcifications were not associated mosis and preterm birth, low birth weight, and small size for
with an increased incidence of neurological sequelae. Choriore- gestational age [68]. Freeman et al. reported that infected babies
tinitis was the main ocular sequelae, found in almost all children were born earlier than uninfected babies and that congenital
and noted years after birth, despite specific therapy in the first infection was associated with an increased risk of preterm deliv-
year of life. ery when seroconversion occurred before 20 weeks of gestation.
An important step in the diagnosis of congenital toxoplasmo- Congenital infection was not associated with low birth weight
sis and evaluation of time of infection is achieved by laboratory or small size for gestational age. The cause for shorter gestation
techniques, monitoring the immune response: titer and affin- is not yet known. The highest frequency of severe abnormalities
ity of specific antibodies (Fig. 1). Other laboratory tools focus at birth is seen in children whose mothers acquired a primary
on direct detection of the parasite by animal or tissue inocula- infection between the 10th and 24th week of gestation [67]. The
tion or more commonly, by molecular techniques. Carvalheiro et likelihood of clinical symptoms in the newborn is reduced when
al. studied the incidence of congenital toxoplasmosis in Brazil, infection occurs later.
based on persistence of anti-Toxoplasma IgG antibodies beyond Clinical manifestations in newborns with congenital toxo-
the age of 1year. Disease incidence was estimated to be 3.3 per plasmosis vary and can develop at different times before and
10,000. A definitive diagnosis was confirmed in five infants with after birth. Most newborns infected with T. gondii are asymp-
both serum IgM and/or IgA antibodies, and clinical abnormali- tomatic at birth (70–90%) [61]. When clinical manifestations are
present they are mainly non-specific and may include: a mac-
ulopapular rash, generalized lymphadenopathy, hepatomegaly,
splenomegaly, hyperbilirubinemia, anemia and thrombocytope-
nia [69]. The classic triad of chorioretinitis, intracranial calcifi-
cations and hydrocephalus is found in fewer than 10% of infected
infants [47]. Hydrocephalus and/or microcephaly may develop
when intra-uterine infection results in meningo-encepahlitis
[69]. All these signs and symptoms are included in the general
work-up of suspected congenital TORCH infections: toxoplas-
mosis, other (syphilis, varicella-zoster, parvovirus B19), rubella,
cytomegalovirus (CMV) and herpes infections. Cerebral cal-
cifications can be demonstrated by cranial radiography, ultra-
sonography or computerized tomography. Neurologic impair-
ment may initially present as seizures, necessitating specific
evaluation and treatment.
The most prevalent consequence of congenital toxoplasmosis
is chorioretinitis.
Chorioretinitis is diagnosed based on characteristic retinal
infiltrates. Vutova et al. [70] investigated eye manifestations of
congenital toxoplasmosis in 38 infants and children. The most
Fig. 1. Laboratory diagnosis of congenital toxoplasmosis. frequent finding was chorioretinitis (92%), together with other
E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472 463

ocular lesions in 71% of cases, and the second most common sue inoculation or more commonly, by molecular techniques. It
finding was microphthalmia with strabismus. Lesions of the is important to combine all available clinical and laboratory data
anterior segment of the eye included iridocyclitis, cataracts and during the evaluation of toxoplasmosis diagnosis and providing
glaucoma. Other uncommon findings were diminished visual treatment recommendations.
acuity and neurological sequelae such as hydrocephalus, calci- Infection during gestation may cause serious damage to
fication in the brain, paresis, and epilepsy. the fetus and hence, a major objective of the diagnosis is to
Wallon et al. [71] reported the clinical evolution of ocular estimate the time of maternal infection. IgG antibodies usu-
lesions and final visual function, in a prospective cohort of 327 ally appear within two weeks of infection, peak within 6–8
congenitally infected children in France. The children were iden- weeks and persist in the body indefinitely [67]. IgM antibod-
tified by maternal prenatal screening and monitored for up to ies are considered the indicators of recent infection and can
14 years. After 6 years, 79 (24%) children had at least one be detected by enzyme immunoassay (EIA) or immunosor-
retinochoroidal lesion. In 23 of them a new lesion was diag- bent agglutination assay test (IAAT) relatively early—within
nosed within10 years, mainly in a previously healthy location. 2 weeks of infection. Uncertainty may arise as IgM may per-
Normal vision was found in about two thirds of children with sist for years following primary infection [73]. IgA antibodies
lesions in one eye, half the children with lesions in both eyes may also persist for more than a year [67] and their detection
and none had bilateral visual impairment. Most of the mothers is informative mainly for the diagnosis of congenital toxoplas-
(84%) had been treated. A combination of pyrimethamine and mosis. The level of specific IgE antibodies increases rapidly
sulfadiazine had been prescribed in all the children (38% before and remains detectable for less than 4 months after infection,
and 72% after birth). Late-onset retinal lesions and relapse can which leaves a very short time to be used for diagnostic pur-
occur many years after birth, but the overall ocular prognosis of poses [74]. However, IgE serology is not useful in samples from
congenital toxoplasmosis seems satisfactory, when infection is newborns.
identified early and appropriately treated. Early diagnosis and When serology alone is insufficient direct evidence for
treatment are believed to reduce the risk of visual impairment. toxoplasma infection should be sought. Both the laboratory
Relevant laboratory tests include complete blood count performing the tests and the referring physician should be
(CBC), liver function tests and specific T. gondii diagnostic tests aware of the limitations and select the best combination of
as described in details below. If T. gondii infection is suspected tests available to timely evaluate the stage of toxoplasma
at the time of birth, diagnostic work-up includes ophthalmic, infection [75]. Laboratory tests available are summarized in
auditory and neurological examinations, lumbar puncture and Table 1.
cranial imaging [69].
In a large percentage of children the disease sequelae may 4.3.1. Sabin Feldman dye test (SFDT)
become apparent and present with visual impairment, mental and This is the first test developed for the laboratory diagnosis of
cognitive abnormalities of variable severity, seizures or learning T. gondii infection [76], it is still considered the “gold standard”.
disabilities only after several months or years [55]. SFDT detects the presence of anti-T. gondii specific antibod-
Infants born to women infected simultaneously with HIV ies (total Ig) and is performed only in reference centers. The
and T. gondii should be evaluated for congenital toxoplasmosis, change in antibody titer as determined in SFDT in consecu-
considering the increased risk of reactivation of parasitemia and tive serum samples taken at least 3 weeks apart is important
disease in these mothers. for the evaluation of infection during pregnancy. A “significant”
In a case–control study in Israel, Potasman et al. tested 95 change is considered to be at least a four-fold difference. The
children with variable neurological disorders: cerebral palsy, absolute antibody titer is also important—values over 250 IU/ml
epilepsy and nerve deafness compared with a control group of are considered “high” suggestive of recent infection. The tested
109 healthy children, for the presence of T. gondii-specific anti- sera are serially diluted and incubated with live tachyzoites
bodies in the serum. They found that children with any of the (carrying toxoplasma-specific antigens) in the presence of sep-
neurological disorders were significantly more likely to have arated human plasma from “sero-negative” donors (providing
T. gondii specific IgG antibodies, especially those with nerve complement components). The antigen–antibody–complement
deafness (relative risk 2.5 and 7.1, respectively) [72]. complexes formed are subsequently lysed in the presence of
A definite diagnosis cannot be made in the following situa- the dye methylene blue. End-point titer is established by count-
tions: (1) the infant is older than one year of age and was not ing the numbers of dead (unstained) and live (stained) para-
tested for toxoplasmosis previously, (2) either the child or the sites. The reported titer is that producing lysis of 50% of the
mother is seronegative, or (3) the mother was known to be sero- organisms. End-point titer can be converted to international
positive prior to conception. units (IU): additional standardization is achieved by prepara-
tion of a standardised control serum (consisting of a pool of
4.3. Prenatal laboratory diagnosis sera), tested by numerous reference centers, and adjusted so
that the SFDT value of this control serum is set at 1000 IU/ml
The principle method used to diagnose and evaluate tim- [77]. Recently, the WHO recognized the first international stan-
ing of congenital infection relies on indirect evidence, and is dard for human anti-toxoplasma IgG, with an assigned potency
based on detection of specific antibodies, by monitoring the of 20 IU per ampoule of total anti-toxoplasma antibodies
immune response. Direct evidence is obtained by animal or tis- [78].
464 E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472

Table 1
Laboratory diagnostic tests for congenital toxoplasmosis
Test Matrix Results Interpretation Time Degree of Other remarks Suggested use
expertise
equired

Sabin Feldman Dye Serum Titer in international Quantitative data: Routine = ∼2–4 High, reference Gold standard Confirmation of
Test (SFDT) units (IU) of total detection of high × week center only infection
specific Ig (≥250 IU) antibodies
titers and significant
changes (≥×4) in
titer in consecutive
samples – important
for evaluation of
recent infection
“hands Live parasites
on” = several and animal
hours injection → risk
to lab employee
Standardized Follow-up
assay change in titer
(international
effort)
EIAa /total Igb Serum Positive/negative for Exposure to T. gondii Several hours Low = simple Possible false Screening test
total specific Ig automated test negative very
early infection
IFAc -total Ig Serum Titer (in IU) of total Exposure to T. gondii Several hours High, reference Very When SFDT is
specific Ig center only subjective and unavailable
difficult to
standardize
IgG by EIA Serum Positive/negative for Exposure to T. gondii Several hours Low = simple Partial results Screening test
specific IgG Abs automated test (combine with
IgM detection)
IgM/IgA or IgE by Serum Positive/negative for Possible recent Several hours Low = simple Requires IgM –
EIA specific IgM, IgA or infection with T. automated test further testing, Screening
IgE Abs gondii IgE – not in
newborn
IgM/IgA –
Newborn
IgE – Earlier
IgM/IgA or IgE by Serum Positive/negative for Possible recent Several hours Relatively high Most sensitive IgM/IgA –
IAAT specific IgM, IgA or Infection with T. and specific Newborn
IgE Abs gondii test
Western blot
should be
considered if
contamination
with maternal
blood is
suspected
IgM/IgA or IgE by Serum Positive/negative for Possible recent Several hours High, reference Very When ISAGA is
IFA specific IgM, IgA or Infection with T. center only subjective and unavailable
IgE Abs gondii difficult to
standardize
IgG avidity Serum Avidity = functional High avidity supports Several hours Relatively Supportive When only a
affinity “past infection” (≥4 simple evidence single serum
months) sample is
available, in the
beginning of
pregnancy
Mice Body flu- Positive/negative Presence of parasite 3–6 weeks High, reference Low Strain isolation
ids/tissue center only sensitivity
Live parasites
and animal
injection → risk
to lab employee
E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472 465

Table 1 (Continued)
Test Matrix Results Interpretation Time Degree of Other remarks Suggested use
expertise
equired

Cells Body flu- Positive/negative Presence of parasite 3–6 days Very high Low When available
ids/tissue reference center sensitivity for a direct proof
only of infection
Live parasites
PCR Body flu- Positive/negative Presence of parasite’s Several hours High High Amniotic fluid
ids/tissue DNA sensitivity
Western blot IgG, Serum Identical/unidentical Fetal/newborn 1 day High, reference Infrequent Confirmatory
IgM to maternal Ig infection center availability test or
fetal/newborn
infection
a EIA: enzyme immunoassay.
b Ig: immunoglobulin.
c IFA: indirect fluorescent assay.

4.3.2. Enzyme immunoassays (EIA) genital infection (where the expected levels of antibodies are
The most common laboratory tests for toxoplasmosis very low) [74,83].
infection, also available as commercial kits and/or auto- Toxoplasma-specific IgE antibodies can be detected by EIA
mated platforms, are EIA. These tests include: enzyme-linked or IAAT in sera of recently infected adults, congenitally infected
immunosorbent assay (ELISA) and enzyme linked fluorescent infants, and children with congenital toxoplasmic chorioretinitis
immuno-assay (ELFA) which test for the presence of IgG and/or [84]. IgE detection is, however, ineffective in evaluating fetal or
IgM antibodies specific for the parasite in human sera. EIA are newborn samples where IgA tests are most informative.
useful as fast, low-cost screening tests and have been improved
over the years to avoid false positive results due to non-specific 4.3.4. Indirect fluorescent assay (IFA)
detection of interfering factors such as rheumatoid factor and The IFA was widely used to demonstrate T. gondii-specific
antinuclear antibodies. antibodies: serially diluted serum samples are incubated with
There is no standardization of these tests, which causes high live, inactivated toxoplasma fixed to a glass slide. T. gondii-
variability in results obtained with different kits and/or in differ- specific antibodies present in the serum would bind to the inacti-
ent laboratories. Consequently, and also as a result of the high vated parasite, and the complex is then detected using fluorescein
incidence of false-positive results even in reference centers, the isothiocyanate-labeled anti-human Ig (or anti-IgG or anti-IgM).
US Food and Drug Administration (FDA) issued a health advi- IFA is safer to perform and more economical than the SFDT. It
sory to physicians on July, 1997. The FDA recommends avoiding appears to measure the same antibodies as the dye test, and its
reliance of results obtained with any single commercial kit for titers tend to parallel dye test titers [47,85]. However, the IFA
the detection of toxoplasma-specific IgM, as the sole deter- interpretation is subjective and time consuming. False positive
minant of recent toxoplasma infection in pregnant women. In results may occur with sera containing antinuclear antibodies
our experience at the Israeli National Toxoplasmosis Reference and rheumatoid factor [86], and false negative results of IFA for
Center, during the years 1997–2002, in an average of 747 sam- IgM may occur due to blockage by T. gondii-specific IgG [87].
ples (range: 652–816) received annually for confirmation, only
17% ± 2.6% were indeed positive for T. gondii-specific IgM. It 4.3.5. Avidity
is therefore recommended that patient follow-up would be per- IgG avidity testing was developed by Hedman et al. and is
formed by a reference center, and that commercial kits would be based on the increase in functional affinity (avidity) between
locally evaluated to achieve the highest degree of accuracy and T. gondii-specific IgG and the antigen over time, as the host
repeatability possible for screening tests. immune response (and specific B cell selection) evolves [88].
In general, when toxoplasma infection is suspected based on Dissociation of the antigen–antibody complexes reflects the
detection of specific IgM antibodies specimens are referred for lower avidity closer to primary infection. Pregnant women with
confirmation by a reference center where SFDT, PCR and other high avidity antibodies are those who have been infected at least
advanced assays can be performed. 3–5 months earlier, which makes the avidity test most useful and
reliable in the first trimester when high-avidity is detected [89].
4.3.3. Immunosorbent agglutination assay test (IAAT) In one study, 35 out of 63 patients (55%) who were classified
IAAT is highly specific in detection of anti-T. gondii IgM, by toxoplasma-specific serology as having recent or border-
IgA or IgE antibodies [79]. This assay utilizes the entire tachy- line infection showed high avidity-antibodies and were therefore
zoite and is the most sensitive commercially available method treated as chronic patients [90]. Lappaplainen et al. [91] were
[80–82]. Unfortunately, it is expensive, requires a high degree of able to follow 13 women who showed high-avidity antibodies in
expertise and is not automated. It is consequently seldom used in the first trimester and confirmed that none of the born infants was
reference centers, usually in neonates suspected of having con- found to be infected with T. gondii (as determined serologically
466 E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472

after birth). The avidity test is most important when only a sin- As in all diagnostic tests based on amplification of DNA,
gle serum sample is available at the time when critical decisions a few technical aspects are of crucial importance in achieving
must be made. To the best of our knowledge, commercial IgG reliable results. Therefore, PCR based test should be carefully
avidity kits have been licensed in Europe but not in the US [92]. designed to include negative, positive and internal control, target
When avidity is low or borderline it may be misleading and DNA for amplification should be specific, sample preparation
a more careful interpretation of all laboratory tests results in techniques should be perfected to extract minute parasite DNA
conjunction with other clinical findings, should then be under- [105] and to prevent cross contamination.
taken. Several studies have shown that this test is reliable and In a small (5 laboratories) inter-laboratories comparative
valuable in diagnosis of recent infection during early pregnancy work [106] followed by a larger study (15 laboratories) [104] sig-
[88,93–98]. nificant differences in test performances were obtained, includ-
Accurate and definitive serologic diagnosis of recently ing false negatives and false positives. These results should
acquired toxoplasma infection is still difficult and depends on definitely urge optimization and standardization of the test. More
testing of more than one sample. Efforts to develop better recently, three PCR protocols were optimized prior to a compar-
diagnostic approaches continue based on antigens specifically ative study, using three different targets: 18S ribosomal DNA,
expressed either during the primary phase (i.e. GRA7, GRA4) B1 gene and AF146527. No significant difference was observed
or the latent phase (i.e. GRA1) of infection. These antigens can between the results of the three protocols [107].
be produced by recombinant DNA technologies and may lead to Chabbert et al. [108] used two different primer sets of the B1
a more informative serologic diagnosis, based on a single serum gene to compare PCR performance followed by Southern blot,
sample [47,99,100]. on various sample types (including amniotic fluid, blood and
tissues). For amniotic fluid both PCR conditions produced sim-
4.3.6. Animal and cell culture inoculation ilar results. The fragments produced by one of the primer sets
A definite laboratory confirmation of active toxoplasmosis had to be confirmed by specific hybridization, otherwise non-
infection (especially in immunocompromised patients and preg- specific results were obtained. The PCR product of the same
nant women) can be established by inoculation of body fluids or amplification procedure was sequenced by Kompalic-Cristo and
tissue into mice or cell culture [47]. suspected of originating from human DNA, as predicted by
Mice are injected intraperitoneally or subcutaneously with bioinformatics analysis [109].
10–30 ml of sediment from amniotic fluid or whole fetal blood. Different protocols influence the sensitivity and specificity
The mice are bled prior and 3–6 weeks following inoculation. of PCR assays. The specificity and positive predictive value of
Antibody detection by SFDT establishes infection and final PCR tests on amniotic fluid samples is close to 100% [110,111].
proof is obtained by staining to demonstrate brain cysts [101]. However, the sensitivity of these PCR tests varies and estimated,
Cell culture inoculation with amniotic fluid or blood uses based on a large number of studies, to be 70–80% [105]. One
indirect IFA to detect the parasite in monolayers within 3–6 report showed that the sensitivity of PCR from amniotic fluid is
days following inoculation [102]. When compared, inoculation affected by the stage of pregnancy in which maternal infection
of both blood and amniotic fluid from an infected fetus resulted occurs: best sensitivity was detected when maternal infection
in toxoplasma isolation from both cultures in 70% of cases. occurred between 17 and 21 weeks of pregnancy [89,111,112].
However, in 40% of the cases T. gondii isolation is successful in In addition, treatment with anti-toxoplasma drugs may also
only one of the samples [100]. Derouin et al. [100] demonstrated affect the sensitivity [89,112]. However, the reliability of a PCR
similar sensitivities comparing cell culture and mice inoculation. test performed on amniotic fluid prior to the 18th week of preg-
Thulliez et al. [102] reported that the sensitivity of amniotic nancy requires further evaluation [110,111]. It should also be
fluid cell culture inoculation is only 53% compared with 73% noted, that testing amniotic fluid for T. gondii was found to be
sensitivity in mice inoculation. effective about 4 weeks following infection, which is already
Currently, the principle role for these methods may be con- during the parasitemic stage in the infected mother. Therefore,
firmation of PCR as they are complex, expensive and relatively PCR test should not be performed in the absence of serologic or
insensitive. [103]. other clinical/sonographic data indicative of infection.
In the last 4 years there have been reports on the use of Real
4.3.7. Molecular diagnosis Time PCR, a sensitive and specific technique, which enables
Replacing fetal blood analysis, which is a high risk proce- rapid detection of amplification products as well as hybridization
dure for the fetus, with molecular evaluation of amniotic fluid of amplicon-specific probes, similar to PCR followed by South-
has provided a low risk diagnosis of congenital toxoplasmosis. ern blot analysis. The method, which will ultimately replace
Polymerase chain reaction (PCR) is currently the most common traditional PCR, enables an overall time for amplification and
molecular technique routinely used for diagnosis of toxoplasmo- detection of less than two hours. In addition, cross contamina-
sis, although, it has not yet been standardized. No attempts have tion is prevented by elimination of the need to handle amplified
been made to standardize either the sample preparation process amplicons. In Real Time PCR it is possible to perform a quanti-
or the PCR amplification itself, and numerous laboratories use tative study and follow the parasite load, allowing determination
multiple “in-house” methods of varying sensitivities and relia- of parasite count and its correlation with clinical symptoms and
bility [104,105]. Recently, a commercial PCR proficiency test impact of treatment. The technique permits linear range over 6
became available. logs of DNA concentrations [113,114].
E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472 467

The most popular target gene for PCR diagnosis of T. gondii drugs such as spiramycin (adult dosage 3–4 g/d × 3–4 weeks)
is the 35-fold repetitive gene B1. A variety of primers have been and sometimes clindamycin are recommended in certain circum-
used for amplification, some of which include nested primers. stances. Spiramycin is used to prevent placental infection; it is
The second common locus is the single copy gene P30 also used in many European countries especially France, Asia and
known as SAG1, which encodes for a surface antigen. Another South America. In the US, spiramycin is currently not approved
PCR target is the 18S ribosomal DNA. As reviewed by Bastien by the FDA but, available as an investigational drug, requiring
[105], two other target loci have been examined but are currently special approval. Treatment with pyrimethamine and sulfadi-
not used by most laboratories. Recently, some laboratories have azine to prevent fetal infection is contraindicated during the first
shown success in amplification of a DNA fragment, AF146527, trimester of pregnancy due to concerns regarding teratogenicity,
which is repeated 200–300 times [89,113,114]. except when the mother’s health is seriously endangered. During
the first trimester sulfadiazine can be used alone.
4.4. Laboratory diagnosis of infants As recently reviewed by Montoya and Liesenfeld [112], treat-
ment protocols vary among different centers. The effectivity of
Laboratory diagnosis of Toxoplasma infection in infants is anti-T. gondii treatment is evaluated based on two criteria: rate
based on a combination of serologic tests, parasite isolation, of mother to child transmission and prevalence and severity of
and nonspecific findings [112]. sequelae. The majority of the studies are retrospective or cohort
When suspected, serologic follow-up of the newborn is rec- studies of various populations and case definitions. The differ-
ommended for the first year of life [90]. Evaluation for direct ence in study patterns and methodologies affects the reliability
evidence as described above should be repeated as well during and validity of the results and thus prevents issuing further rec-
this period. ommendations.
Serologic tests should follow total (or IgG) T. gondii specific Wallon et al. [120] reviewed studies comparing treated and
antibodies titer (taking into account that closely after birth these untreated concurrent groups of pregnant women with proved
are maternal in origin, transferred through the placenta), IgM or likely acute toxoplasma infection. Outcomes data of the
and IgA titers. Though passively transferred maternal IgG has a offspring were reported. The results showed treatment to be
half life of approximately 1 month, it can still be detected in the effective in five studies but ineffective in four. Gras et al. [121]
newborn for several months, generally disappearing completely reported that the effect of prenatal pyrimethamine–sulfadiazine
within one year [112]. Appearance of autonomous IgG antibod- combination treatment on the cerebral and ocular sequelae of
ies in a congenitally infected newborn begins, in an untreated intrauterine infection with T. gondii was not beneficial in 181
patient, about 3 months after birth. Anti-parasitic therapy may children of infected mothers. Neto reported the outcome of
delay antibody production for about 6 months and, occasionally, patients with congenital toxoplasmosis who were all treated with
may completely prevent antibodies production [86]. pyrimethamine, sulfadiazine and folinic acid; of 195 patients 138
(71%) were asymptomatic until the age of 2 years. The authors
4.4.1. Western blots suggest that for six patients with sequelae because of the delay
Remington et al. introduced Western blots (using T. gondii- in anti-toxoplasma treatment (6–14 months post diagnosis) the
specific labeled antigens to detect antibodies, separated by elec- disease was not prevented [122]. Gratzl et al. [123] reported vari-
trophoresis and transferred to a membrane) to compare newborn able concentrations of spiramycin and its metabolites in serum
versus maternal antibodies [115–117]. Western blotting could and amniotic fluid of 18 pregnant women following treatment.
potentially separate maternal from fetal/newborn antibodies. All the drug concentrations were below the level reported to
The test is not widely used mainly because of its technical com- inhibit parasite growth in vitro. The authors suggested that the
plexity and high price. possible reasons being individual pharmatokinetic variability
and patients’ treatment compliance. Gilbert et al. [124] reported
5. Treatment of congenital toxoplasmosis the effect of prenatal treatment in 554 infected women and
their offspring. In this study comparison of early versus late
Anti T. gondii treatment initiation generally requires con- treatment and of combination treatment (pyrimethamine, sulfa-
firmatory laboratory tests in a reference center, followed by diazine) with spiramycin or no-treatment, were all statistically
consultation with experts. Treatment is indicated in the fol- insignificant. The possible interpretation is that delayed treat-
lowing conditions: infection during pregnancy and congenital ment initiation led to failure to prevent parasite transmission.
infection as well as infection of an immunocompromised host Another European multicenter study comparing transmission
(e.g. HIV/AIDS) and in case of an invasive disease. In pregnant rates and clinical outcomes in 856 mother–infant pairs, found
women and infected neonates, both symptomatic and asymp- no significant association between the outcome and the intensity
tomatic, specific treatment of T. gondii infection is indicated of treatment protocol in pregnancy [125]. Bessieres et al. [126]
immediately following established diagnosis. The combination studied the effect of treatment during pregnancy in a cohort of
of pyrimethamine, (adult dosage 25–100 mg/d × 3–4 weeks), 165 women and found that cases could be identified during preg-
sulfadiazine adult dosage 1–1.5 g qid × 3–4 weeks) and folinic nancy as well as during the neonatal period. They also noted
acid (leucovorin, 10–25 mg with each dose of pyrimethamine, that T. gondii was less frequently isolated in women treated
to avoid bone marrow suppression) is the basic treatment pro- with pyrimethamine and sulfadoxine than in women treated
tocol recommended by the WHO [118] and CDC [119]. Other with spiramycin only. Foulon et al. [127] reviewed the measures
468 E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472

of prevention of congenital toxoplasmosis and concluded that State of Goias, Brazil as recommended by experts [127]. Screen-
treatment during pregnancy significantly reduces sequelae and ing of women should begin prior to conception with follow-up
treatment of infected children has a beneficial effect when ther- monthly tests during pregnancy to detect seroconverion. This is
apy is begun soon after birth. the basis for the French [138] screening program and the Aus-
In conclusion, the efficacy of anti-T. gondii treatment in preg- trian Toxoplasmosis Prevention Programs, both recommend rou-
nancy is still an unsettled matter. It is difficult to find the effect tine serologic testing, in Austria three times during pregnancy:
of treatment when comparing the different studies because of: in the first, second and third trimesters and in France six times
different treatment regimes and timing (for small groups of following the initial finding [139]. Treatment is recommended
patients), the pharmacokinetics patterns of drugs (concentra- if one of the tests suggests definite or probable primary mater-
tion in amniotic fluid and fetal CSF), patient (none) compliance nal infection [140]. In Massachusetts, USA, where there is low
with treatment and different methodologies of follow-up in each seroprevalence in the population, only newborns are screened
study. for the presence of T. gondii-specific IgM [141]. IgM detec-
As concluded by Peyron et al. [128] and others, further large tion is followed by an extensive clinical evaluation and a one
scale, carefully controlled studies are necessary in order to clar- year treatment regimen combination of pyrimethamine and sul-
ify this controversial issue. At present the anti-parasite treatment fadiazine [140]. A recent study screened 364,130 neonates in
recommended for toxoplasmosis as outlined above, should be the United States for T. gondii specific IgM and confirmed 195
considered as the guideline for good medical practice. cases of congenital toxoplasmosis (1 in 1867). Moreover, a 7-
year follow-up of the treated patients revealed no symptoms or
6. Prevention at least no progress of the disease. Based on these findings, the
authors suggest including toxoplasmosis in neonatal screening
6.1. Primary prevention programs [122].
In the United Kingdom a national committee concluded that
In the United States efforts at prevention of congenital tox- no prenatal or neonatal screening for T. gondii should be per-
oplasmosis have been primarily directed towards health educa- formed, which brought out controversy among specialists [142].
tion, focused to avoid personal exposure to the parasite (hygienic A survey conducted in Italy reported 35/1000 pregnant
and culinary practice during pregnancy). In Poland, an extensive women with primary T. gondii infection and recommended
health education campaign, increased toxoplasmosis awareness maternal screening during pregnancy rather than neonatal
and knowledge of preventive behaviour significantly within the screening [143]. In Norway, screening of pregnant women was
4 years of the reported study [129]. Many other countries have recommended until 1977 when the National Institute of Public
introduced educational programs aimed at reducing the inci- Health discouraged it, following a large study that showed low
dence of congenital toxoplasmosis. Such programs depend on (0.17 %) incidence of primary infection during pregnancy [144].
careful identification of unique target-populations and tailoring Two years following this change in policy, a study by Eskild et
appropriate approaches of education. To evaluate the success of al. [145] showed that despite the recommendations, 81% of the
such programs it is important to measure incidence rate before pregnant women were still routinely tested for T. gondii-specific
onset and at pre-determined intervals after introducing the cam- antibodies.
paign. In Finland, a cost-benefit analyses of screening programs for
pregnant women as well as education programs revealed the ben-
6.1.1. Vaccine eficial effect of such programs in both low and high incidences
Development of a vaccine for toxoplasmosis can prevent of toxoplasmosis [146].
human disease by immunization of human as well as animals Cost-effectiveness of optional screening programs (no
(the source of infection). Both attenuated parasite and immuno- screening, pre-conception or neonates screening, frequency of
genic antigens are considered as potential agents for vaccination. tests during pregnancy) depends on local factors: incidence of
Live attenuated S48 strain is in use for vaccination of sheep in congenital toxoplasmosis, available diagnostic and therapeutic
Europe and New Zealand but is unsuitable for human use due to services, and the population compliance with screening. It is
its expense, short shelf life and most importantly, to the ability of important to promote public, as well as professional, knowledge
the attenuated parasite to revert to a pathogenic strain [130–133]. regarding the disease, in order to effectively prevent, diagnose
Much of the work has been focused on SAG1, a surface anti- and treat congenital toxoplasmosis.
gen expressed on tachyzoites, in attempts to induce protective In conclusion, it is highly recommended to educate the pub-
immune response (mainly T-helper response) when introduced lic and professionals to minimize risk of infection. Screening
to the host with various adjuvants [134–136]. Development of programs of women at childbearing age and upon gestation or
vaccine using antigens expressed by bradyzoites and oocytes is at least newborn screening is highly effective for early treatment
also under investigation[134,137]. and prevention of sequelae.

6.2. Secondary prevention – screening Acknowledgments

Routine toxoplasmosis screening programs for pregnant Dr. Irena Volovik Sub-district Health Officer, Hadera, Israel,
women have been established in France, in Austria and in the for providing data of the presented case.
E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472 469

Mrs. R. Kaufman and Mrs. R. Avni for excellent laboratory [23] Barragan A, Sibley LD. Transepithelial migration of Toxoplasma
work. gondii is linked to parasite motility and virulence. J Exp Med
2002;195:1625–33.
[24] Su C, Howe DK, Dubey JP, Ajioka JW, Sibley LD. Identification of
quantitative trait loci controlling acute virulence in Toxoplasma gondii.
References Proc Natl Acad Sci USA 2002;99:10753–8.
[25] Weiss LM, Kim K. The International Congress on Toxoplasmosis. Int
[1] Dubey JP, Lindsay DS, Speer CA. Structures of Toxoplasma gondii J Parasitol 2004;34:249–52.
tachyzoites, bradyzoites, and sporozoites and biology and development [26] Kissinger JC, Gajria B, Li L, Paulsen IT, Roos DS. ToxoDB: accessing
of tissue cysts. Clin Microbiol Rev 1998;11:267–99. the Toxoplasma gondii genome. Nucleic Acids Res 2003;31:234–6.
[2] Black MW, Boothroyd JC. Lytic cycle of Toxoplasma gondii. Microbiol [27] Bhopale GM. Pathogenesis of toxoplasmosis. Comp Immunol Micro-
Mol Biol Rev 2000;64:607–23. biol Infect Dis 2003;26:213–22.
[3] Lekutis C, Ferguson DJ, Boothroyd JC. Toxoplasma gondii: identifica- [28] Sibley LD, Boothroyd JC. Virulent strains of Toxoplasma gondii com-
tion of a developmentally regulated family of genes related to SAG2. prise a single clonal lineage. Nature 1992;359:82–5.
Exp Parasitol 2000;96:89–96. [29] Howe DK, Sibley LD. Toxoplasma gondii comprises three clonal lin-
[4] Lyons RE, McLeod R, Roberts CW. Toxoplasma gondii tachyzoite- eages: correlation of parasite genotype with human disease. J Infect
bradyzoite interconversion. Trends Parasitol 2002;18:198–201. Dis 1995;172:1561–6.
[5] Manger ID, Hehl AB, Boothroyd JC. The surface of Toxoplasma [30] Ware PL, Kasper LH. Strain-specific antigens of Toxoplasma gondii.
tachyzoites is dominated by a family of glycosylphosphatidylinositol- Infect Immun 1987;55:778–83.
anchored antigens related to SAG1. Infect Immun 1998;66:2237–44. [31] Kong JT, Grigg ME, Uyetake L, Parmley S, Boothroyd JC. Serotyping
[6] Dubey JP, Miller NL, Frenkel JK. The Toxoplasma gondii oocyst from of Toxoplasma gondii infections in humans using synthetic peptides. J
cat feces. J Exp Med 1970;132:636–62. Infect Dis 2003;187:1484–95.
[7] Coutinho SG, Lobo R, Dutra G. Isolation of Toxoplasma from the [32] Ajzenberg D, Cogne N, Paris L, Bessieres MH, Thulliez P, Filisetti
soil during an outbreak of toxoplasmosis in a rural area in Brazil. J D, et al. Genotype of 86 Toxoplasma gondii isolates associated with
Parasitol 1982;68:866–8. human congenital toxoplasmosis, and correlation with clinical findings.
[8] Bahia-Oliveira LM, Jones JL, Azevedo-Silva J, Alves CC, Orefice F, J Infect Dis 2002;186:684–9.
Addiss DG. Highly endemic, waterborne toxoplasmosis in north Rio [33] Ferguson DJ, Pittilo RM. Toxoplasma gondii and the professor. Para-
de Janeiro state, Brazil. Emerg Infect Dis 2003;9:55–62. sitol Today 1999;15:301–2.
[9] Benenson MW, Takafuji ET, Lemon SM, Greenup RL, Sulzer AJ. [34] Grigg ME, Bonnefoy S, Hehl AB, Suzuki Y, Boothroyd JC. Success
Oocyst-transmitted toxoplasmosis associated with ingestion of contam- and virulence in Toxoplasma as the result of sexual recombination
inated water. N Engl J Med 1982;307:666–9. between two distinct ancestries. Science 2001;294:161–5.
[10] Bowie WR, King AS, Werker DH, Isaac-Renton JL, Bell A, Eng [35] Su C, Evans D, Cole RH, Kissinger JC, Ajioka JW, Sibley LD. Recent
SB, et al. Outbreak of toxoplasmosis associated with municipal expansion of Toxoplasma through enhanced oral transmission. Science
drinking water. The BC Toxoplasma Investigation Team. Lancet 2003;299:414–6.
1997;350:173–7. [36] Volkman SK, Hartl DL, Parasitology. A game of cat and mouth. Sci-
[11] Dumetre A, Darde ML. How to detect Toxoplasma gondii oocysts in ence 2003;299:353–4.
environmental samples? FEMS Microbiol Rev 2003;27:651–61. [37] Suzuki Y, Wong SY, Grumet FC, Fessel J, Montoya JG, Zolopa AR,
[12] Jones JL, Lopez A, Wilson M, Schulkin J, Gibbs R. Congenital toxo- et al. Evidence for genetic regulation of susceptibility to toxoplasmic
plasmosis: a review. Obstet Gynecol Surv 2001;56:296–305. encephalitis in AIDS patients. J Infect Dis 1996;173:265–8.
[13] Hoff EF, Carruthers VB. Is Toxoplasma egress the first step in inva- [38] Mack DG, Johnson JJ, Roberts F, Roberts CW, Estes RG, David C, et
sion? Trends Parasitol 2002;18:251–5. al. HLA-class II genes modify outcome of Toxoplasma gondii infec-
[14] Barragan A, Sibley LD. Migration of Toxoplasma gondii across bio- tion. Int J Parasitol 1999;29:1351–8.
logical barriers. Trends Microbiol 2003;11:426–30. [39] Herwaldt BL. Laboratory-acquired parasitic infections from accidental
[15] Kasper LH, Mineo JR. Attachment and invension of host cell by Tox- exposures. Clin Microbiol Rev 2001;14:659–88, table of contents.
oplasma gondii. Parasitol Today 1994;10:82–5. [40] Tenter AM, Heckeroth AR, Weiss LM. Toxoplasma gondii: from ani-
[16] Suss-Toby E, Zimmerberg J, Ward GE. Toxoplasma invasion: the mals to humans. Int J Parasitol 2000;30:1217–58.
parasitophorous vacuole is formed from host cell plasma mem- [41] Jones JL, Kruszon-Moran D, Wilson M, McQuillan G, Navin T,
brane and pinches off via a fission pore. Proc Natl Acad Sci USA McAuley JB. Toxoplasma gondii infection in the United States: sero-
1996;93:8413–8. prevalence and risk factors. Am J Epidemiol 2001;154:357–65.
[17] Hakansson S, Morisaki H, Heuser J, Sibley LD. Time-lapse video [42] Cook AJ, Gilbert RE, Buffolano W, Zufferey J, Petersen E, Jenum
microscopy of gliding motility in Toxoplasma gondii reveals a PA, et al. Sources of toxoplasma infection in pregnant women: Euro-
novel, biphasic mechanism of cell locomotion. Mol Biol Cell pean multicentre case–control study. European Research Network on
1999;10:3539–47. Congenital Toxoplasmosis. BMJ 2000;321:142–7.
[18] Mordue DG, Desai N, Dustin M, Sibley LD. Invasion by Toxoplasma [43] Aspinall TV, Marlee D, Hyde JE, Sims PF. Prevalence of Toxoplasma
gondii establishes a moving junction that selectively excludes host cell gondii in commercial meat products as monitored by polymerase chain
plasma membrane proteins on the basis of their membrane anchoring. reaction—food for thought? Int J Parasitol 2002;32:1193–9.
J Exp Med 1999;190:1783–92. [44] Lappin MR, Burney DP, Hill SA, Chavkin MJ. Detection of Toxo-
[19] Mordue DG, Hakansson S, Niesman I, Sibley LD. Toxoplasma gondii plasma gondii-specific IgA in the aqueous humor of cats. Am J Vet
resides in a vacuole that avoids fusion with host cell endocytic and Res 1995;56:774–8.
exocytic vesicular trafficking pathways. Exp Parasitol 1999;92:87–99. [45] Dubey JP. Duration of immunity to shedding of Toxoplasma gondii
[20] Lecordier L, Mercier C, Sibley LD, Cesbron-Delauw MF. Transmem- oocysts by cats. J Parasitol 1995;81:410–5.
brane insertion of the Toxoplasma gondii GRA5 protein occurs after [46] Dubey JP. Sources of Toxoplasma gondii infection in pregnancy. Until
soluble secretion into the host cell. Mol Biol Cell 1999;10:1277–87. rates of congenital toxoplasmosis fall, control measures are essential.
[21] Opitz C, Soldati D. ’The glideosome’: a dynamic complex power- BMJ 2000;321:127–8.
ing gliding motion and host cell invasion by Toxoplasma gondii. Mol [47] Dubey JP, Beattie CP. Toxoplasmosis of animals and man. Boca Raton:
Microbiol 2002;45:597–604. CRC Press; 1988.
[22] Sibley LD. Intracellular parasite invasion strategies. Science [48] Franklin DM, Dror Z, Nishri Z. The prevalence and incidence of Tox-
2004;304:248–53. oplasma antibodies in pregnant women. Isr J Med Sci 1993;29:285–6.
470 E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472

[49] Teutsch SM, Juranek DD, Sulzer A, Dubey JP, Sikes RK. Epi- [71] Wallon M, Kodjikian L, Binquet C, Garweg J, Fleury J, Quantin C, et
demic toxoplasmosis associated with infected cats. N Engl J Med al. Long-term ocular prognosis in 327 children with congenital toxo-
1979;300:695–9. plasmosis. Pediatrics 2004;113:1567–72.
[50] Stagno S, Dykes AC, Amos CS, Head RA, Juranek DD, Walls K. [72] Potasman I, Davidovitch M, Tal Y, Tal J, Zelnik N, Jaffe M. Congenital
An outbreak of toxoplasmosis linked to cats. Pediatrics 1980;65:706– toxoplasmosis: a significant cause of neurological morbidity in Israel?
12. Clin Infect Dis 1995;20:259–62.
[51] Isaac-Renton J, Bowie WR, King A, Irwin GS, Ong CS, Fung CP, et [73] Bobic B, Sibalic D, Djurkovic-Djakovic O. High levels of IgM anti-
al. Detection of Toxoplasma gondii oocysts in drinking water. Appl bodies specific for Toxoplasma gondii in pregnancy 12 years after
Environ Microbiol 1998;64:2278–80. primary toxoplasma infection. Case report. Gynecol Obstet Invest
[52] Kourenti C, Heckeroth A, Tenter A, Karanis P. Development and appli- 1991;31:182–4.
cation of different methods for the detection of Toxoplasma gondii in [74] Pinon JM, Toubas D, Marx C, Mougeot G, Bonnin A, Bonhomme A,
water. Appl Environ Microbiol 2003;69:102–6. et al. Detection of specific immunoglobulin E in patients with toxo-
[53] Mead PS, Slutsker L, Dietz V, McCaig LF, Bresee JS, Shapiro C, et plasmosis. J Clin Microbiol 1990;28:1739–43.
al. Food-related illness and death in the United States. Emerg Infect [75] Roberts A, Hedman K, Luyasu V, Zufferey J, Bessieres MH, Blatz
Dis 1999;5:607–25. RM, et al. Multicenter evaluation of strategies for serodiagnosis of
[54] Lynfield R, Guerina NG. Toxoplasmosis. Pediatr Rev 1997;18:75–83. primary infection with Toxoplasma gondii. Eur J Clin Microbiol Infect
[55] Feigen RD, Adcock LM, Edwards MS. Fungel and protozoal infec- Dis 2001;20:467–74.
tions (Toxoplasmosis). In: Fanaroff AA, Martin RJ, editors. Neonatal [76] Sabin AB, Feldman HA. Dyes as microchemical indicators of a new
perinatal medicine disease of the fetus and infant. 5th ed. Mosby Year immunity phenomenon affecting a protozon parasite (Toxoplasma). Sci-
Book; 1992. p. 688–90. ence 1948;108:660–3.
[56] Guerina NG. Congenital infection with Toxoplasma gondii. Pediatr [77] Reiter-Owona I, Petersen E, Joynson D, Aspock H, Darde ML,
Ann 1994;23:138–42, 47–51. Disko R, et al. The past and present role of the Sabin-Feldman dye
[57] Avelino MM, Campos Jr D, do Carmo Barbosa de Parada J, de Castro test in the serodiagnosis of toxoplasmosis. Bull World Health Org
AM. Pregnancy as a risk factor for acute toxoplasmosis seroconversion. 1999;77:929–35.
Eur J Obstet Gynecol Reprod Biol 2003;108:19–24. [78] Rigsby P, Rijpkema S, Guy EC, Francis J, Das RG. Evaluation
[58] Dollfus H, Dureau P, Hennequin C, Uteza Y, Bron A, Dufier JL. of a candidate international standard preparation for human anti-
Congenital toxoplasma chorioretinitis transmitted by preconceptionally Toxoplasma immunoglobulin G. J Clin Microbiol 2004;42:5133–8.
immune women. Br J Ophthalmol 1998;82:1444–5. [79] Stepick-Biek P, Thulliez P, Araujo FG, Remington JS. IgA antibodies
[59] Silveira C, Ferreira R, Muccioli C, Nussenblatt R, Belfort Jr R. Toxo- for diagnosis of acute congenital and acquired toxoplasmosis. J Infect
plasmosis transmitted to a newborn from the mother infected 20 years Dis 1990;162:270–3.
earlier. Am J Ophthalmol 2003;136:370–1. [80] Duffy KT, Wharton PJ, Johnson JD, New L, Holliman RE. Assessment
[60] Chemla C, Villena I, Aubert D, Hornoy P, Dupouy D, Leroux B, et al. of immunoglobulin-M immunosorbent agglutination assay (ISAGA) for
Preconception seroconversion and maternal seronegativity at delivery detecting toxoplasma specific IgM. J Clin Pathol 1989;42:1291–5.
do not rule out the risk of congenital toxoplasmosis. Clin Diagn Lab [81] Ashburn D, Joss AW, Pennington TH, Ho-Yen DO. Specificity and
Immunol 2002;9:489–90. usefulness of an IgE immunosorbent agglutination assay for toxoplas-
[61] Lebech M, Joynson DH, Seitz HM, Thulliez P, Gilbert RE, Dutton mosis. J Clin Pathol 1995;48:64–9.
GN, et al. Classification system and case definitions of Toxoplasma [82] Desmonts G, Naot Y, Remington JS, Immunoglobulin. M-
gondii infection in immunocompetent pregnant women and their con- immunosorbent agglutination assay for diagnosis of infectious diseases:
genitally infected offspring. European Research Network on Congen- diagnosis of acute congenital and acquired Toxoplasma infections. J
ital Toxoplasmosis. Eur J Clin Microbiol Infect Dis 1996;15:799– Clin Microbiol 1981;14:486–91.
805. [83] Wong SY, Hajdu MP, Ramirez R, Thulliez P, McLeod R, Remington
[62] El Ayoubi M, de Bethmann O, Monset-Couchard M. Lenticulostriate JS. Role of specific immunoglobulin E in diagnosis of acute toxo-
echogenic vessels: clinical and sonographic study of 70 neonatal cases. plasma infection and toxoplasmosis. J Clin Microbiol 1993;31:2952–9.
Pediatr Radiol 2003;33:697–703. [84] Remington PL, Anderson DE, Manering MC, Peterson EA, Anderson
[63] Gay-Andrieu F, Marty P, Pialat J, Sournies G, Drier de Laforte T, H. The PRECEDES Project: background and methods. Wis Med J
Peyron F. Fetal toxoplasmosis and negative amniocentesis: necessity 1990;89:695–6.
of an ultrasound follow-up. Prenat Diagn 2003;23:558–60. [85] Araujo FG, Handman E, Remington JS. Use of monoclonal antibodies
[64] Crino JP. Ultrasound and fetal diagnosis of perinatal infection. Clin to detect antigens of Toxoplasma gondii in serum and other body fluids.
Obstet Gynecol 1999;42:71–80, quiz 174–5. Infect Immun 1980;30:12–6.
[65] Safadi MA, Berezin EN, Farhat CK, Carvalho ES. Clinical presentation [86] Wilson M, Jones JL, McAuley JB. Toxoplasma. In: Murray PR, Baron
and follow up of children with congenital toxoplasmosis in Brazil. Braz EJ, Jorgensen JH, Pfaller MA, Yolken RH, editors. Manuel of clinical
J Infect Dis 2003;7:325–31. microbiology, Vol. 2, 8th ed. ASM Press; 2003. p. 1970–80.
[66] Carvalheiro CG, Mussi-Pinhata MM, Yamamoto AY, De Souza CB, [87] Remington JS, Araujo FG, Desmonts G. Recognition of different Tox-
Maciel LM. Incidence of congenital toxoplasmosis estimated by neona- oplasma antigens by IgM and IgG antibodies in mothers and their
tal screening: relevance of diagnostic confirmation in asymptomatic congenitally infected newborns. J Infect Dis 1985;152:1020–4.
newborn infants. Epidemiol Infect 2005;133:485–91. [88] Montoya JG, Liesenfeld O, Kinney S, Press C, Remington JS. VIDAS
[67] Remington JS, Desmonts G. Infectious Diseases of the Fetus and New- test for avidity of Toxoplasma-specific immunoglobulin G for confir-
born Infant. In: Remington JS, Klein JD, editors. Toxoplasmosis. 3rd matory testing of pregnant women. J Clin Microbiol 2002;40:2504–8.
ed. Philadelphia: WB Saunders; 1990. p. 89–195. [89] Remington JS, Thulliez P, Montoya JG. Recent developments for diag-
[68] Freeman K, Oakley L, Pollak A, Buffolano W, Petersen E, Sem- nosis of toxoplasmosis. J Clin Microbiol 2004;42:941–5.
prini AE, et al. Association between congenital toxoplasmosis and [90] Montoya JG. Laboratory diagnosis of Toxoplasma gondii infection and
preterm birth, low birthweight and small for gestational age birth. toxoplasmosis. J Infect Dis 2002;185(Suppl. 1):S73–82.
BJOG 2005;112:31–7. [91] Lappalainen M, Koskiniemi M, Hiilesmaa V, Ammala P, Teramo K,
[69] Swisher CN, Boyer K, McLeod R. Congenital toxoplasmosis. The Tox- Koskela P, et al. Outcome of children after maternal primary Toxo-
oplasmosis Study Group. Semin Pediatr Neurol 1994;1:4–25. plasma infection during pregnancy with emphasis on avidity of specific
[70] Vutova K, Peicheva Z, Popova A, Markova V, Mincheva N, Todorov T. IgG. The Study Group. Pediatr Infect Dis J 1995;14:354–61.
Congenital toxoplasmosis: eye manifestations in infants and children. [92] Alvarado-Esquivel C, Sethi S, Janitschke K, Hahn H, Liesen-
Ann Trop Paediatr 2002;22:213–8. feld O. Comparison of two commercially available avidity tests
E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472 471

for toxoplasma-specific IgG antibodies. Arch Med Res 2002;33: [113] Homan WL, Vercammen M, De Braekeleer J, Verschueren H. Iden-
520–3. tification of a 200- to 300-fold repetitive 529 bp DNA fragment in
[93] Liesenfeld O, Montoya JG, Kinney S, Press C, Remington JS. Effect of Toxoplasma gondii, and its use for diagnostic and quantitative PCR.
testing for IgG avidity in the diagnosis of Toxoplasma gondii infection Int J Parasitol 2000;30:69–75.
in pregnant women: experience in a US reference laboratory. J Infect [114] Reischl U, Bretagne S, Kruger D, Ernault P, Costa JM. Comparison
Dis 2001;183:1248–53. of two DNA targets for the diagnosis of Toxoplasmosis by real-time
[94] Pietkiewicz H, Hiszczynska-Sawicka E, Kur J, Petersen E, Nielsen PCR using fluorescence resonance energy transfer hybridization probes.
HV, Stankiewicz M, et al. Usefulness of Toxoplasma gondii-specific BMC Infect Dis 2003;3:7.
recombinant antigens in serodiagnosis of human toxoplasmosis. J Clin [115] Robert-Gangneux F, Commerce V, Tourte-Schaefer C, Dupouy-Camet
Microbiol 2004;42:1779–81. J. Performance of a Western blot assay to compare mother and newborn
[95] Li S, Galvan G, Araujo FG, Suzuki Y, Remington JS, Parmley S. anti-Toxoplasma antibodies for the early neonatal diagnosis of con-
Serodiagnosis of recently acquired Toxoplasma gondii infection using genital toxoplasmosis. Eur J Clin Microbiol Infect Dis 1999;18:648–
an enzyme-linked immunosorbent assay with a combination of recom- 54.
binant antigens. Clin Diagn Lab Immunol 2000;7:781–7. [116] Pinon JM, Dumon H, Chemla C, Franck J, Petersen E, Lebech M, et
[96] Aubert D, Maine GT, Villena I, Hunt JC, Howard L, Sheu M, al. Strategy for diagnosis of congenital toxoplasmosis: evaluation of
et al. Recombinant antigens to detect Toxoplasma gondii-specific methods comparing mothers and newborns and standard methods for
immunoglobulin G and immunoglobulin M in human sera by enzyme postnatal detection of immunoglobulin G, M, and A antibodies. J Clin
immunoassay. J Clin Microbiol 2000;38:1144–50. Microbiol 2001;39:2267–71.
[97] Li S, Maine G, Suzuki Y, Araujo FG, Galvan G, Remington JS, et al. [117] Tissot Dupont D, Fricker-Hidalgo H, Brenier-Pinchart MP, Bost-Bru
Serodiagnosis of recently acquired Toxoplasma gondii infection with C, Ambroise-Thomas P, Pelloux H. Usefulness of Western blot in sero-
a recombinant antigen. J Clin Microbiol 2000;38:179–84. logical follow-up of newborns suspected of congenital toxoplasmosis.
[98] Nigro M, Gutierrez A, Hoffer AM, Clemente M, Kaufer F, Carral L, Eur J Clin Microbiol Infect Dis 2003;22:122–5.
et al. Evaluation of Toxoplasma gondii recombinant proteins for the [118] Drugs used in parasitic diseases, 2nd ed. Geneva: World Health Orga-
diagnosis of recently acquired toxoplasmosis by an immunoglobulin G nization; 1995.
analysis. Diagn Microbiol Infect Dis 2003;47:609–13. [119] Chin J. Toxoplasmosis. In: Control of Communicable Disease Manual.
[99] Daffos F, Forestier F, Capella-Pavlovsky M, Thulliez P, Aufrant C, 17th ed. Washington DC: American Public Health Association; 2000.
Valenti D, et al. Prenatal management of 746 pregnancies at risk for p. 500–3.
congenital toxoplasmosis. N Engl J Med 1988;318:271–5. [120] Wallon M, Liou C, Garner P, Peyron F. Congenital toxoplasmosis:
[100] Derouin F, Mazeron MC, Garin YJ. Comparative study of tissue cul- systematic review of evidence of efficacy of treatment in pregnancy.
ture and mouse inoculation methods for demonstration of Toxoplasma BMJ 1999;318:1511–4.
gondii. J Clin Microbiol 1987;25:1597–600. [121] Gras L, Gilbert RE, Ades AE, Dunn DT. Effect of prenatal treatment
[101] Derouin F, Thulliez P, Candolfi E, Daffos F, Forestier F. Early prenatal on the risk of intracranial and ocular lesions in children with congenital
diagnosis of congenital toxoplasmosis using amniotic fluid samples and toxoplasmosis. Int J Epidemiol 2001;30:1309–13.
tissue culture. Eur J Clin Microbiol Infect Dis 1988;7:423–5. [122] Neto EC. Newborn screening for congenital infectious diseases. Emerg
[102] Thulliez P, Daffos F, Forestier F. Diagnosis of Toxoplasma infection Infect Dis 2004;10:1068–73.
in the pregnant woman and the unborn child: current problems. Scand [123] Gratzl R, Sodeck G, Platzer P, Jager W, Graf J, Pollak A, et al. Treat-
J Infect Dis Suppl 1992;84:18–22. ment of toxoplasmosis in pregnancy: concentrations of spiramycin and
[103] Foulon W, Pinon JM, Stray-Pedersen B, Pollak A, Lappalainen M, neospiramycin in maternal serum and amniotic fluid. Eur J Clin Micro-
Decoster A, et al. Prenatal diagnosis of congenital toxoplasmosis: a biol Infect Dis 2002;21:12–6.
multicenter evaluation of different diagnostic parameters. Am J Obstet [124] Gilbert RE, Gras L, Wallon M, Peyron F, Ades AE, Dunn DT. Effect
Gynecol 1999;181:843–7. of prenatal treatment on mother to child transmission of Toxoplasma
[104] Pelloux H, Guy E, Angelici MC, Aspock H, Bessieres MH, Blatz gondii: retrospective cohort study of 554 mother-child pairs in Lyon,
R, et al. A second European collaborative study on polymerase chain France. Int J Epidemiol 2001;30:1303–8.
reaction for Toxoplasma gondii, involving 15 teams. FEMS Microbiol [125] Gilbert R, Dunn D, Wallon M, Hayde M, Prusa A, Lebech M, et
Lett 1998;165:231–7. al. Ecological comparison of the risks of mother-to-child transmission
[105] Bastien P. Molecular diagnosis of toxoplasmosis. Trans R Soc Trop and clinical manifestations of congenital toxoplasmosis according to
Med Hyg 2002;96(Suppl 1):S205–15. prenatal treatment protocol. Epidemiol Infect 2001;127:113–20.
[106] Guy EC, Pelloux H, Lappalainen M, Aspock H, Hassl A, Melby KK, [126] Bessieres MH, Berrebi A, Rolland M, Bloom MC, Roques C, Cassaing
et al. Interlaboratory comparison of polymerase chain reaction for the S, et al. Neonatal screening for congenital toxoplasmosis in a cohort
detection of Toxoplasma gondii DNA added to samples of amniotic of 165 women infected during pregnancy and influence of in utero
fluid. Eur J Clin Microbiol Infect Dis 1996;15:836–9. treatment on the results of neonatal tests. Eur J Obstet Gynecol Reprod
[107] Filisetti D, Gorcii M, Pernot-Marino E, Villard O, Candolfi E. Diag- Biol 2001;94:37–45.
nosis of congenital toxoplasmosis: comparison of targets for detection [127] Foulon W, Naessens A, Ho–Yen D. Prevention of congenital toxoplas-
of Toxoplasma gondii by PCR. J Clin Microbiol 2003;41:4826–8. mosis. J Perinat Med 2000;28:337–45.
[108] Chabbert E, Lachaud L, Crobu L, Bastien P. Comparison of two widely [128] Peyron F, Wallon M, Liou C, Garner P. Treatments for toxoplasmosis
used PCR primer systems for detection of toxoplasma in amniotic fluid, in pregnancy. Cochrane Database Syst Rev 2000:CD001684.
blood, and tissues. J Clin Microbiol 2004;42:1719–22. [129] Pawlowski ZS, Gromadecka-Sutkiewicz M, Skommer J, Paul M,
[109] Kompalic-Cristo A, Nogueira SA, Guedes AL, Frota C, Gonzalez LF, Rokossowski H, Suchocka E, et al. Impact of health education on
Brandao A, et al. Lack of technical specificity in the molecular diag- knowledge and prevention behavior for congenital toxoplasmosis: the
nosis of toxoplasmosis. Trans R Soc Trop Med Hyg 2004;98:92–5. experience in Poznan, Poland. Health Educ Res 2001;16:493–502.
[110] Hohlfeld P, Daffos F, Costa JM, Thulliez P, Forestier F, Vidaud M. [130] Letscher-Bru V, Pfaff AW, Abou-Bacar A, Filisetti D, Antoni E, Vil-
Prenatal diagnosis of congenital toxoplasmosis with a polymerase chain lard O, et al. Vaccination with Toxoplasma gondii SAG–1 protein is
reaction test on amniotic fluid. N Engl J Med 1994;331:695–9. protective against congenital toxoplasmosis in BALB/c mice but not
[111] Romand S, Wallon M, Franck J, Thulliez P, Peyron F, Dumon H. in CBA/J mice. Infect Immun 2003;71:6615–9.
Prenatal diagnosis using polymerase chain reaction on amniotic fluid [131] Haumont M, Delhaye L, Garcia L, Jurado M, Mazzu P, Daminet
for congenital toxoplasmosis. Obstet Gynecol 2001;97:296–300. V, et al. Protective immunity against congenital toxoplasmosis with
[112] Montoya JG, Liesenfeld O. Toxoplasmosis Lancet 2004;363:1965– recombinant SAG1 protein in a guinea pig model. Infect Immun
76. 2000;68:4948–53.
472 E. Rorman et al. / Reproductive Toxicology 21 (2006) 458–472

[132] Nielsen HV, Lauemoller SL, Christiansen L, Buus S, Fomsgaard A, [140] Lopez A, Dietz VJ, Wilson M, Navin TR, Jones JL. Prevent-
Petersen E. Complete protection against lethal Toxoplasma gondii ing congenital toxoplasmosis. MMWR Recomm Rep 2000;49:59–
infection in mice immunized with a plasmid encoding the SAG1 gene. 68.
Infect Immun 1999;67:6358–63. [141] Hsu HW, Grady GF, Maguire JH, Weiblen BJ, Hoff R. Newborn
[133] Denkers EY, Gazzinelli RT. Regulation and function of T-cell-mediated screening for congenital Toxoplasma infection: five years experi-
immunity during Toxoplasma gondii infection. Clin Microbiol Rev ence in Massachusetts USA. Scand J Infect Dis Suppl 1992;84:59–
1998;11:569–88. 64.
[134] Bhopale GM. Development of a vaccine for toxoplasmosis: current [142] Gilbert RE, Peckham CS. Congenital toxoplasmosis in the United
status. Microbes Infect 2003;5:457–62. Kingdom: to screen or not to screen? J Med Screen 2002;9:135–41.
[135] Roque-Resendiz JL, Rosales R, Herion P. MVA ROP2 vaccinia virus [143] Ricci M, Pentimalli H, Thaller R, Rava L, Di Ciommo V. Screening
recombinant as a vaccine candidate for toxoplasmosis. Parasitology and prevention of congenital toxoplasmosis: an effectiveness study in
2004;128:397–405. a population with a high infection rate. J Matern Fetal Neonatal Med
[136] Mohamed RM, Aosai F, Chen M, Mun HS, Norose K, Belal US, et al. 2003;14:398–403.
Induction of protective immunity by DNA vaccination with Toxoplasma [144] Eskild A, Magnus P. Commentary: Little evidence of effective pre-
gondii HSP70, HSP30 and SAG1 genes. Vaccine 2003;21:2852–61. natal treatment against congenital toxoplasmosis––the implications for
[137] Buxton D, Innes EA. A commercial vaccine for ovine toxoplasmosis. testing in pregnancy. Int J Epidemiol 2001;30:1314–5.
Parasitology 1995;110(Suppl):S11–6. [145] Eskild A, Fallas Dahl G, Melby KK, Nesheim BI. Testing for toxo-
[138] Thulliez P. Screening programme for congenital toxoplasmosis in plasmosis in pregnancy: a study of the routines in primary antenatal
France. Scand J Infect Dis Suppl 1992;84:43–5. care. J Med Screen 2003;10:172–5.
[139] Aspock H, Pollak A. Prevention of prenatal toxoplasmosis by serolog- [146] Lappalainen M, Sintonen H, Koskiniemi M, Hedman K, Hiilesmaa V,
ical screening of pregnant women in Austria. Scand J Infect Dis Suppl Ammala P, et al. Cost-benefit analysis of screening for toxoplasmosis
1992;84:32–7. during pregnancy. Scand J Infect Dis 1995;27:265–72.

Вам также может понравиться