Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

CLINICAL MICROBIOLOGY REVIEWS, Apr. 2003, p. 265–272 Vol. 16, No.

2
0893-8512/03/$08.00⫹0 DOI: 10.1128/CMR.16.2.265–272.2003
Copyright © 2003, American Society for Microbiology. All Rights Reserved.

Toxocariasis: Clinical Aspects, Epidemiology, Medical Ecology,


and Molecular Aspects
Dickson Despommier*
Department of Environmental Health Sciences and Department of Microbiology, Mailman School
of Public Health, Columbia University, New York, New York 10032

INTRODUCTION .......................................................................................................................................................265
HISTORY OF DISCOVERY .....................................................................................................................................265
LIFE CYCLE ...............................................................................................................................................................266
CLINICAL ASPECTS.................................................................................................................................................267
CLINICAL SIGNS AND SYMPTOMS ....................................................................................................................268
VLM ..........................................................................................................................................................................268
OLM .........................................................................................................................................................................269
DIAGNOSIS ................................................................................................................................................................269
TREATMENT..............................................................................................................................................................269
EPIDEMIOLOGY .......................................................................................................................................................269
MEDICAL ECOLOGY...............................................................................................................................................270
MOLECULAR ASPECTS ..........................................................................................................................................270
CONCLUDING REMARKS ......................................................................................................................................271
REFERENCES ............................................................................................................................................................271

INTRODUCTION ocular larva migrans (OLM), in which toxocariasis pathological


effects on the host are restricted to the eye and the optic nerve.
Toxocariasis is the clinical term applied to infection in the
T. canis and T. cati are distributed worldwide, due to human
human host with either Toxocara canis or Toxocara cati. Both
settlement of nearly all land masses. Our penchant (almost a
of these are ascarid nematodes in the order Ascaridida, super-
genetic imperative) for surrounding ourselves with various do-
family Ascaridiodea, family Toxocaridae. Their definitive hosts
mestic animals, particularly cats and dogs, has ensured a world-
are the domestic dog and cat, in which they live as adults within
wide distribution for toxocariasis. In addition, many places that
the lumen of the small intestine. Infection can occur by the
remain permanently uninhabited also have robust populations
host ingesting viable, embryonated eggs from contaminated
of feral dogs and cats, introduced there during the 17th
sources (e.g., soil and earthworms, etc.), or they can acquire
through the 19th centuries by sea-faring nations, thus facilitat-
the infection in utero (i.e., transplacentally) from the infected
ing the maintenance of these two ascarid infections on numer-
mother when she ingests more infective eggs. In contrast, the
ous islands throughout the Pacific Ocean basin, including the
human host is aberrant with respect to the completion of the
Galapagos Islands.
life cycle. Infective larvae hatch after ingestion of eggs, but the
Several species of Toxocara infecting domestic cats and
juvenile stages fail to develop to mature adult worms. Instead,
other felids have been recently identified. Toxocara malaya-
they wander throughout the body for months or up to several
siensis n. sp. (18) infects domestic cats, while Toxocara lyncus
years, causing damage to whatever tissue they happen to enter.
infects the caracal (30). It is not known whether either of these
The ability of a eukaryotic parasite to survive in any mammal
newly described entities cause human infection resulting in
for that length of time is unusual. Only a few others have
clinical disease, but given the epidemiological pattern estab-
evolved long-term survival strategies; namely, the adult stage
lished by T. canis and T. cati, it is likely that at least a portion
of schistosomes live for 10 to 25 years, the first-stage larva of
of the total number of aberrant infections in humans have
Trichinella spiralis lives for the life span of the host, some
resulted from exposure to their migrating larvae. This review
species of adult filarial nematodes live 10 to 15 years, and the
concentrates on the most prevalent of Toxocara species, name-
juvenile stage of most species of tapeworms survive for 5 to 10
ly, T. canis and T. cati.
years. To accomplish this daunting feat, all of these parasites
have acquired unique mechanisms for evading the host’s im-
mune system. Toxocara spp. are no exception. HISTORY OF DISCOVERY
The dominant clinical manifestations associated with toxo-
cariasis are classified according to the organs affected. There Human infection with Toxocara spp. was first described by
are two main syndromes; visceral larva migrans (VLM), which Wilder (61) in 1950. He identified a nematode larva of un-
encompasses diseases associated with the majors organs, and known species within a retinal granuloma of a child. In 1952,
Beaver and colleagues (4) reported on a similar series of chil-
dren who presented with high circulating eosinophilia, and
* Mailing address: Department of Environmental Health Sciences,
Mailman School of Public Health, Columbia University, 722 W. 168th
suffered from severe, long-term, multisystem disease. From
St., New York, NY 10032. Phone: (212) 781-6670. Fax: (212) 781-1830. this group of patients, they described most of the clinical fea-
E-mail: ddd1@columbia.edu. tures of VLM and, in histopathological sections of tissues ob-

265
266 DESPOMMIER CLIN. MICROBIOL. REV.

FIG. 1. Life cycle of T. canis and T. cati.

tained at biopsy, correctly classified the causative agents as the eggs. This situation arises from indiscriminate defecation on
larva of either T. canis or T. cati. Since that time, the juveniles these sites by cats and dogs that harbor the adult worms. After
of these two parasite species have been detected in a variety of ingestion, the eggs hatch to release larvae (juveniles) that pen-
lesions of the eye and throughout the body (31, 59) in patients etrate the small intestine, enter the circulation, and then are
from all corners of the world. Today, the public health com- free to wander throughout the body, with the possibility of
munity at-large acknowledges that toxocariasis in all its clinical invading all organs. There is controversy as to whether these
forms constitutes a major health risk, especially among chil- are second- or third-stage larvae. In the definitive host, the
dren exhibiting pica. juvenile parasites go on to complete the life cycle, which, in
many ways, resembles that of Ascaris lumbricoides, the ascarid
infecting humans. Transplacental infection is common in both
LIFE CYCLE
dogs and cats. In addition, the definitive host can become
Ingestion of the embryonated eggs of Toxocara (Fig. 1) ini- infected by ingesting embryonated eggs carried by paratenic
tiates infection in both the definitive and aberrant host. Chil- hosts, such as earthworms, ants, and other soil-dwelling inver-
dren accidentally come into contact with them when they play tebrates. Humans can develop aberrant infections by ingesting
in sandboxes and on playgrounds contaminated with Toxocara these same animals.
VOL. 16, 2003 TOXOCARIASIS 267

FIG. 2. Portion of dog small intestine with adult T. canis. Male worms have a curled tail; females have a straight tail.

Worms develop to the adult stage (Fig. 2) in the small retina, inducing granulomatous reactions (Fig. 7; Fig. 8) lead-
intestine about 60 to 90 days after the larvae hatch. Mating ing to impaired sight. In severe cases, the granuloma was re-
then ensues, giving rise to nonembryonated eggs (Fig. 3), which sponsible for the loss of sight. These pathological manifesta-
become excreted with the fecal mass. Embryonation occurs in tions have, in the past, occasionally been misdiagnosed as
the soil within a week or so after deposition (Fig. 4). Incuba- retinoblastoma (28, 62). Today, with reliable immunodiagnos-
tion periods of longer duration are due to lower temperatures. tic reagents and methods, OLM is almost never mistaken for
In northern latitudes, eggs can lie dormant until the tempera- other clinical entities. Epidemiologic evidence suggests that
ture rises in spring, triggering embryonation. ocular disease tends to occur in the absence of systemic in-
volvement and vice versa, which has led to the proposal that
CLINICAL ASPECTS the two manifestations of this infection be reclassified as OLM

The degree of host damage, and the concomitant elicitation


of signs and symptoms, varies with regard to which tissue has
been invaded; the liver (20), lungs (27), and central nervous
system (CNS) (32), including the eyes (49), appear to be most
sensitive. In addition, the number of migrating juveniles and
the age of the host are two additional factors important as to
whether a given individual’s condition will become elevated
above the clinical horizon. Pathological consequences are
largely dependent upon the death of the juveniles. Their death
heralds the onset of marked delayed-type and immediate-type
hypersensitivity responses. Inflammation manifests as eosino-
philic granulomas. The immediate hypersensitivity responses
to dying and dead larvae in the viscera, including the lungs,
liver (Fig. 5), and brain (Fig. 6), produce symptoms character-
istic of VLM.
In the eye, migrating third stage larvae can damage the FIG. 3. Unembryonated egg of T. canis (90 by 75 ␮m).
268 DESPOMMIER CLIN. MICROBIOL. REV.

FIG. 4. Embryonated egg of T. canis (90 by 75 ␮m).

and VLM (19). It is possible that there are strains of T. canis


with specific tropisms. Alternatively, VLM may reflect the con-
sequences of a host inflammatory response to repeated waves
FIG. 6. Juvenile T. canis in liver of an experimentally infected
of migrating larvae through the viscera, whereas OLM occurs
mouse.
in individuals who have not been previously sensitized (24).

CLINICAL SIGNS AND SYMPTOMS ranges in spectrum from asymptomatic infection to larvae mi-
VLM grating in specific target organs (38, 52, 57). In the lungs, larval
migrations may result in asthma (6, 56). T. canis has been
VLM is mainly a disease of young children (⬍5 years old) suggested as an environmental risk factor for asthma among
(63). It presents with fever; enlargement and necrosis of the some inner-city populations (42). Similarly, in the brain, T.
liver (46); enlargement of the spleen; lower respiratory symp- canis has been implicated as one of the causes of so-called
toms (particularly bronchospasm, resembling asthma); eo- idiopathic seizure disorders (9), as well as a cause of functional
sinophilia sometimes approaching 70% (1); and hypergam- intestinal disorders (25). One study implicated Toxocara as a
maglobulinemia of immunoglobulin M (IgM), IgG, and IgE contributing factor in skin disorders of at least two varieties
classes. In the last of these instances, symptoms are more (prurigo and urticaria) (22), while another presented indirect
pronounced, with increased levels of IgE/anti-IgE immune evidence linking Toxocara infection with a form of eosinophilic
complexes (39). Myocarditis (44), nephritis (53), and involve- arthritis (45). In experimental infections in mice, learning be-
ment of the CNS have been described. CNS involvement can havior and memory are affected, and both appear to be dose
lead to seizures, neuropsychiatric symptoms, or encephalopa- and time dependent (8). It is therefore reasonable to speculate
thy. that similar phenomena are likely to be at work in long-term
There is an increasing appreciation that more subtle clinical infections in humans, as well.
manifestations might also arise as a result of long-term expo- Experimental infections in mice have also shed some light as
sure to the migrating juveniles. So-called covert toxocariasis to the effects of VLM on the overall pattern of immune re-
sponses. Inbred C3H/HeN mice infected with T. canis juveniles
had altered patterns of cytokine responses that were presumed

FIG. 5. Juvenile T. canis from impression smear of brain tissue


from an experimentally infected mouse (390 by 30 ␮m). FIG. 7. Portion of retina from a child suffering from OLM.
VOL. 16, 2003 TOXOCARIASIS 269

to favor the survival of the parasite. Both IL-12 and tumor


necrosis factor alpha were significantly lowered in the infected
group compared to controls (26). IL-5 was associated with re-
sistance to Nippostrongylus braziliensis but had no effect against
the migrating larvae of T. canis (11).
Protective immunity is slow to develop, if it develops at all.
This is primarily due to factors most likely related to the ability
of the juvenile to periodically change its antigenic signal. Data
supporting this speculation will be given more attention below.

OLM

OLM usually occurs in children 5 to 10 years old and typi-


cally presents as unilateral vision impairment that is sometimes
accompanied by strabismus (12). The most serious conse-
quence of the infection is invasion of the retina, leading to
granuloma formation, which occurs typically peripherally or in FIG. 8. Portion of a retina from a child suffering from OLM. Note
the posterior pole. These granulomas drag the retina and cre- eosinophilic granulomatous lesion due to reaction against a dead ju-
venile worm.
ate a distortion, heteropia, or detachment of the macula (54).
The degree of visual acuity impairment depends on the specific
area involved, and blindness is common. OLM can also cause TREATMENT
diffuse endophthalmitis or papillitis; secondary glaucoma can
follow. In at least one rare instance following long-term infec- Albendazole is the treatment of choice for toxocariasis. Pa-
tion with Toxocara, a choroidal neovascular membrane formed tients receiving a 5-day treatment course of albendazole (10
after presenting earlier as chorioretinitis (37). mg/kg of body weight/day in two divided doses) improved rel-
ative to patients who received treatment with the older anthel-
minthic drug thiabendazole (55). A dose of 400 mg of al-
DIAGNOSIS bendazole twice a day for 5 days is the currently recommended
therapy (21). Because the other commonly used benzimid-
Any pediatric patient with an unexplained febrile illness azole, mebendazole, is poorly absorbed outside the gastroin-
and eosinophilia should be suspected of having VLM. Hepa- testinal tract, this agent is a second-line treatment, although
tosplenomegaly and evidence of multisystem disease and his- some success has been reported in patients who ingest 1 g or
tory of pica make the diagnosis of VLM more likely. Similarly, more for a 21-day course (21). Symptomatic treatment, includ-
OLM should be suspected in any child with unilateral vision ing administration of corticosteroids, has been helpful for sup-
loss and strabismus. Diagnostic tests for VLM are primarily pressing the intense allergic manifestations of the infection.
immunological (50). The precipitin test is subject to cross- OLM is treated by surgery (vitrectomy), anthelminthic chemo-
reactions with common antigens of the larvae and blood group therapy, and/or corticosteroids (12, 54).
substance A. The enzyme-linked immunosorbent assay (ELISA),
which employs antigens secreted by the second-stage larva, has EPIDEMIOLOGY
sufficient specificity to be the best indirect test for diagnosing
T. canis and T. cati, as alluded to, are unfortunately all too
this infection. Recombinant antigens have been produced from
common parasites of most domestic and peridomestic dogs
the second-stage larvae of T. canis that promises to add even
and cats, particularly young ones. Even those sold through re-
greater specificity to an already-reliable test (approximately
liable kennels and pet shops may harbor adult worms. This is
92%) employing ELISA. The ELISA has a reasonably high because, as stated in the section dealing with its life cycle,
degree of sensitivity, as well (approximately 78%), at a titer puppies and kittens acquire Toxocara juveniles transplacentally
greater than 1:32 (51). from the infected mother. Therefore, having a litter of puppies
Other indicators of infection include hypergammaglobu- in the home has been identified as a significant risk factor (35).
linemia and an elevated isohemagglutinin titer. Thus, a con- As expected, children with pica are at higher risk of ingesting
stellation of clinical disease described above, a history of embryonated eggs from soil than those not exhibiting this be-
pica, eosinophilia, and positive serology, strongly point to the havior. Growing up in a poor neighborhood is associated with
diagnosis. Liver biopsy may reveal a granuloma surrounding a a higher rate of seropositivity for toxocariasis than is being
larva, but a successful diagnosis using this approach is fortu- raised in middle-income bracket housing.
itous at best and not recommended. Outdoor parks in urban and suburban settings are, in most
OLM is diagnosed primarily on the basis of clinical criteria cases, highly contaminated with embryonated eggs of T. canis
during an ophthalmologic examination. The immunodiagnostic and T. cati, since it is in this environment that people routinely
tests used for VLM are not as reliable for OLM. In one study, walk their pets (7, 17, 36, 41, 47). Burgeoning populations of
only 45% of patients with clinically diagnosed OLM had titers urban, semiwild cats and dogs represent a growing problem in
higher than 1:32 (51). many tropical and subtropical regions and most likely contrib-
270 DESPOMMIER CLIN. MICROBIOL. REV.

ute in a major way to the maintenance of high levels of Toxo- Another mechanism for egg dispersal is drinking water. In
cara eggs in the environment (48). It should be added that an one study, public beaches adjacent to municipal drinking water
increased incidence of rabies is also associated with urban and supplies just outside the city limits of Moscow, Russia, were
suburban cohorts of abandoned dogs. Adult patients institu- implicated as a source of contamination (5). The authors spec-
tionalized for mental retardation are also at high risk (23). ulated that by allowing dogs and cats free access to these
Preventing the indiscriminate deposition of dog and cat fe- recreational areas, this increased the likelihood that eggs of
ces in play areas frequented by children appears to be the best Toxocara would enter the water column of the lake. Bathers
control strategy for limiting infection in adolescent populations frequently and inadvertently drink water while wading and
in large urban centers. In one study in Japan, placing clear swimming, allowing for the possibility of ingesting infective
vinyl plastic covers over sandboxes at night seemed to effec- eggs. In addition, this scenario also could lead to eggs contam-
tively discourage pets from using them as fecal dump zones. In inating tap water.
addition, during the summer months, temperatures within the Some elements of soil, saprophytic fungi for example (Pae-
sand often rose above 45°C, a condition created by covering cilomyces lilacinus and Paecilomyces marquandii), have been
them over at night. The authors speculated that this might shown under controlled conditions to have larvicidal activity
render the play area safe for use with respect to Toxocara (60). against the juvenile worm within its egg’s shell (3). However
Routine treatment of nonferal dogs and cats with ivermectin, interesting these findings may be, however, it should be cau-
mebendazole, or other related benzimidazoles is another avail- tioned that their application to the control of Toxocara eggs in
able measure which may prove effective in certain settings to soil may prove intractably difficult, due to the high degree of
limit the spread of this resilient group of parasites. Some cities unpredictability regarding the behavior of species of any kind
have solved the dilemma associated with the use of sandboxes that are introduced into new environments (15).
by eliminating them from municipal parks and playgrounds.
Veterinarians continue to play an important role in combat- MOLECULAR ASPECTS
ing the spread of Toxocara infection in situations where they
see large numbers of dogs and cats that are brought to them by Nematodes in the genus Toxocara are distant relatives of the
pet owners. Recommending regular stool examinations and free-living, soil (and laboratory)-dwelling roundworm Caeno-
the frequent use of chemotherapeutic agents such as mebenda- rhabditis elegans. The latter is the only member of the phylum
zole has proven effective in controlling infection. Nematoda whose entire genomic DNA sequence has been de-
termined (see Nobel Prizes in Medicine and Physiology, 2002).
It contains 19,099 genes (7a) and is 97 Mb in size. The genome
MEDICAL ECOLOGY
of T. canis is approximately the same size as that of A. lumbri-
The physical environment plays a crucial role in maintaining coides, which is about three times larger than that of C. elegans
and distributing the infective eggs of T. canis and T. cati, (3 ⫻ 109 bp) (33). T. canis has 18 chromosomes, compared to
although this subject remains underappreciated. Nonetheless, 24 for Ascaris.
developing effective control programs may require that this Investigations into the molecular biology of Toxocara have
aspect be known in much greater detail. Infective ascarid eggs mainly focused on the secreted proteins of the migrating juve-
of all species can last for months to years outside the host nile stages. These proteins have proven useful in immunodi-
under optimal conditions, due solely to a resistant outer shell agnosis of VLM, and OLM. Speculation favors these same
composed of ascarosides. This acellular layer enables eggs to proteins in aiding the worm regarding its capacity to evade
withstand various harsh chemicals (e.g., high concentrations of potentially protective immune responses. This idea derives
formalin and various inorganic acids), extreme temperature from the fact that the juvenile stage wanders about the tissues
changes, and various degrees of moisture. Future strategies for for months to years without apparent interference from the
reducing the number of infectious eggs in soil must find novel host. Presumably, the worms eventually die of old age. The fact
ways of breaching the egg shell barrier that protects the im- that many of the excretory-secretory proteins from the juvenile
mature worm from its external environment. stages constitute a family of at least six highly antigenic mucins
Earthworms and small mammals play a role in dispersing (13) associated with the cuticular surface reinforces this con-
eggs from a point source. As Darwin pointed out in his essay cept (29). Secreted mucins temporarily coat the surface of the
“On the Formation Of Vegetable Mould through the Action of worm (43) and are shed into the host periodically (2). It is
Worms with Observations on Their Habits” (10), earthworms thought that this shedding behavior represents an attempt on
dump huge quantities of processed (i.e., partially digested) soil the part of the parasite to confuse the host’s immune system,
onto the surface of the ground, bringing it from depths as great leaving behind it a trail of slime, not unlike that of a snail (34).
as 2 ft. Viable eggs become incorporated into their fecal pellets In this model, the worm periodically switches its secreted an-
and are then subject to random distribution throughout the tigenic identity, thus avoiding harm.
local area by rain events and wind. Peridomestic mammals, Many other novel coding regions for other expressed pro-
such as dogs, cats, squirrels, and chipmunks, play a role simi- teins of the resting (dauer) larva have also been reported,
lar to that of earthworms, albeit less efficient, in the dispersal including those encoding four different C-type lectins, five
of embryonated eggs (14). Birds that feed primarily on the varieties of superoxide dismutase, phosphatidylethanolamine
ground (e.g., pigeons, starlings, and sparrows) can serve as binding protein, prohibin, olfactomedin, aquaporin, three unique
paratenic hosts, carrying eggs from place to place on their feet venom allergen/ASP homologues, and an asparaginyl endo-
and beaks, and may be responsible for depositing eggs in places peptidase. Functions for these gene products await further in-
far from the original source. vestigation. Another interesting group of proteins has been
VOL. 16, 2003 TOXOCARIASIS 271

described from various stages of T. canis and may help explain control program. Finally, more effective single-dose treatment
the mechanism employed by the parasite to migrate through regimens with safer (over-the-counter?) drugs for pediatric
host tissues. Cathepsin-z-like protease genes have been cloned patients would help limit the time of illness, provided of course
and their cDNAs have been sequenced, identifying a cysteine that adequate medical infrastructure is already in place. Killing
protease coding region expressed both in the adult and infec- Toxocara eggs in contaminated soils is seen by most epidemi-
tive larva (16). ologists as a nearly impossible task, but if such a strategy could
Molecular vaccines could prove useful in aiding in the con- be found and safely implemented, vast numbers of acres of
trol of infection in domestic dogs and cats. The search so far now potentially dangerous city landscape could be rendered
has identified the myosins of Toxocara as potential candidates Toxocara-free.
(40). Several fragments of myosin proved highly antigenic
when used in combination in the ELISA for IgG antibodies REFERENCES
resulting from naturally occurring infections. In that study, 1. Arango, C. A. 1998. Visceral larva migrans and the hypereosinophilia syn-
drome. S. Med. J. 91:882–883.
over 85% of the patients previously diagnosed with VLM were 2. Badley, J. E., R. B. Grieve, and D. D. Bowman. 1987. Immune-mediated
positive. To date, however, no molecular vaccines have been adherence of eosinophils to Toxocara canis infective larvae: the role of
described. excretory-secretory antigens. Parasite Immunol. 9:133–143.
3. Basualdo, J. A., M. L. Ciarmela, P. L. Sarmiento, and M. C. Minvielle. 2000.
Ascarid nematodes are well-known for their ability to induce Biological activity of Paecilomyces genus against Toxocara canis eggs. Para-
strong allergic responses, and laboratory investigators working sitol. Res. 86:854–859.
with Ascaris spp. have frequently had to cease research on 4. Beaver, P. C., C. H. Snyde, and G. M. Carrera. 1952. Chronic eosinophilia
due to visceral larva migrans. Pediatrics 9:7–19.
them solely for this reason. Allergens have been partially char- 5. Beer, S. A., G. I. Novosilítsev, and L. I. Melínikova. 1999. The role of the
acterized from Ascaris spp. and constitute a group of lipid- water factor in the dissemination of Toxocara eggs and the spread of toxo-
cariasis in a megalopolis. Parasitology 33:129–135.
binding polypeptides expressed as a large aggregate polypro-
6. Buijs, J., G. Borsboom, and J. J. van Gemund. 1994. Toxocara seropreva-
tein referred to as nematode polyprotein allergens (64). The lence in 5-year old elementary schoolchildren: relation with allergic asthma.
parent nematode polyprotein allergen molecule is typically Am. J. Epidemiol. 140:839–847.
7. Castillo, D., C. Paredes, and C. Zanartu. 2000. Environmental contamina-
secreted as a large polymer and then undergoes digestion at tion with Toxocara spp. eggs in public squares and parks from Santiago,
regular intervals along its length, producing a series of poly- Chile, 1999. Bol. Chil. Parasitol. 55:86–91.
peptides of ca. 15 kDa. Each of these smaller molecules is 7a.C. elegans Genome Consortium. 1998. Genome sequence of Caenorhabditis
elegans: a platform for investigating biology. Science 282:201–208.
structurally related to one of two subgroups, A or B. It is these 8. Cox, D. M., and C. V. Holland. 2001. Relationship between three intensity
smaller subunits of the parent molecule that induce allergic levels of Toxocara canis larvae in the brain and effects on exploration,
responses in a wide variety of mammalian hosts. A group of anxiety, learning and memory in the murine host. J. Helminthol. 75:33–41.
9. Critchley, E. M., S. D. Vakil, and D. N. Hutchinson. 1982. Toxoplasma,
polyprotein allergens have been identified from T. canis, des- Toxocara, and epilepsy. Epilepsia 23:315–323.
ignated TBA-1 (65), and are similar in structure to those al- 10. Darwin, C. 1881. On the formation of vegetable mould through the action of
worms with Observations on their habits. John Murray, London, England.
ready described in Ascaris. 11. Dent, L. A., C. M. Daly, and G. Mayrhofer. 1999. Interleukin-5 transgenic
mice show enhanced resistance to primary infections with Nippostrongylus
braziliensis but not to primary infections with Toxocara canis. Infect. Immun.
CONCLUDING REMARKS 67:989–993.
12. Dinning, W. J., S. H. Gillespie, R. I. Cooling, and R. M. Maizels. 1988.
Toxocariasis remains a problem throughout the world, in- Toxocariasis: a practical approach to management of ocular disease. Eye
ducing multisystem disease in young people. Overcrowding 2:580–582.
13. Doedens, A., A. Loukas, and R. M. Maizels. 2001. A cDNA encoding Tc-
and the inevitable cohabitation of our peridomestic environ- MUC-5, a mucin from Toxocara canis larvae identified by expression screen-
ment by dogs and cats reinforces the transmission cycle in an ing. Acta Trop. 79:211–217.
14. Dubinsky, P., K. Havasivoa-Reiterova, and B. Petko. 1995. Role of small
already-dismal situation in many regions. Public parks and mammals in the epidemiology of toxocariasis. Parasitology 110:187–193.
playgrounds have become zones of disease acquisition, not at 15. Elton, C. S. 1958. The ecology of invasions by plants and animals. University
all fitting their original purpose. At the present time, control of Chicago Press, Chicago, Ill.
16. Falcone, F. H., K. K. Tetteh, and P. Hunt. 2000. The new subfamily of
programs aimed at reducing the number of domestic animals, cathepsin-z-like protease genes includes Tc-cpz-1, a cysteine protease gene
or, at the very least, limiting their access to areas frequented by expressed in Toxocara canis adults and infective stage larvae. Exp. Parasitol.
small children, appear to be effective in a few cases. Control of 94:210–217.
17. Giacometti, A., O. Cirioni, and M. Fortuna. 2000. Environmental and sero-
the spread of dog feces, and to a lesser extent cat feces, also logical evidence for the presence of toxocariasis in the urban area of Ancona,
helps limit exposure. Regular treatment of pets with benzimi- Italy. Eur. J. Epidemiol. 16:1023–1026.
18. Gibbons, L. M., D. E. Jacobs, and R. A. Sani. 2001. Toxocara malaysiensis n.
dazoles helps reduce worm burdens and limits the number of sp. (Nematoda:Ascaridoidea) from domestic cat (Felis catus Linnaeus 1758).
eggs deposited in soil. However, for the most part, these hearty J. Parasitol. 87:660–665.
nematodes appear to be more than holding their own. What is 19. Glickman, L. I., and P. M. Schantz. 1982. Epidemiology and pathogenesis of
zoonotic toxocariasis. Epidemiol. Rev. 10:143–148.
needed in terms of future control programs is the development 20. Hartleb, M., and K. Januszewski. 2001. Severe hepatic involvement in vis-
of radically new approaches, such as effective molecular or ceral larva migrans. Eur. J. Gastroenterol. Hepatol. 13:1245–1249.
DNA-based vaccines that offer the possibility of lifelong pro- 21. Hotez, P. J. 1995. Toxocara canis, p. 683–684. In F. D. Burg, E. R. Wald, J. R.
Ingelfinger, and P. A. Polin (ed.), Gellis and Kaganís current pediatric
tection. Oral baits laced with vaccine would be ideal for dealing therapy, 15th ed. W. B. Saunders Pubs., Philadelphia, Pa.
with semiwild dog and cat populations, an approach similar to 22. Humbert, P., M. Niezborala, and R. Salembier. 2000. Skin manifestations
associated with toxocariasis: a case-control study. Dermatology 210:230–234.
ones that already exist for the large-scale control of rabies in 23. Hummer, D., K. Symon, and I. Groskopf. 1992. Seroepidemiologic study of
wild animal populations. To augment vaccine development, toxocariasis and strongyloidiasis in institutionalized mentally retarded adults.
rapid, highly sensitive and specific diagnostic tests employing Am. J. Trop. Med. Hyg. 46:278–281.
24. Kazacos, K. R. 1991. Visceral and ocular larva migrans. Semin. Vet. Med.
recombinant, antigenic peptides that can be executed in the Surg. (Small Anim.) 6:227–235.
field are essential elements needed to formulate any future 25. Konate, A., O. Duhamel, and D. Basset. 1996. Toxocariasis and functional
272 DESPOMMIER CLIN. MICROBIOL. REV.

intestinal disorders. Presentation of 4 cases. Gastroenterol. Clin. Biol. 20: 45. Rayes, A. A., and J. R. Lambertucci. 2001. Human toxocariasis as a possible
909–911. cause of eosinophilic arthritis. Rheumatology 40:109–110.
26. Kuroda, E., Y. Yoshida, and B. En Shan. 2001. Suppression of macrophage 46. Rayes, A. A., D. Teixeira, J. C. Serufo, V. Nobre, C. M. Antunes, and J. R.
interleukin-12 and tumor necrosis factor-alpha production in mice infected Lambertucci. 2001. Human toxocariasis and pyogenic liver abscess: a possi-
with Toxocara canis. Parasite Immunol. 23:305–311. ble association. Am. J. Gastroenterol. 96:563–566.
27. Kuziemski, K., E. Jassem, and E. Mierzejewska. 1999. Lung manifestation of 47. Ruiz de Ybanez, M. R., M. M. Garijo, and F. D. Alonso. 2001. Prevalence and
visceral larva migration syndrome due to Toxocara canis infection. Pneumo- viability of eggs of Toxocara spp. and Toxascaris leonia in public parks in
nol. Alergol. Pol. 67:554–557. eastern Spain. J. Helminthol. 75:169–173.
28. Lopez-Velez, R., M. Suarez de Figueroa, and L. Gimeno. 1995. Ocular toxo- 48. Ruiz de Ybanez, M. R., M. Garijo, M. Goyena, and F. D. Alonso. 2000.
cariasis or retinoblastoma? Enferm. Infec. Microbiol. Clin. 13:242–245. Improved methods for recovering eggs of Toxocara canis from soil. J. Hel-
29. Loukas, A., M. Hintz, and D. Linder. 2000. A family of genes active during minthol. 74:349–353.
the resting stage of the parasite were cloned and their cDNA sequences 49. Sabrosa, N. A., and E. C. de Souza. 2001. Nematode infections of the eye:
determined. Secreted mucins from the parasitic nematode Toxocara canis toxocariasis and diffuse unilateral subacute neuroretinitis. Curr. Opin. Oph-
bears diverse mucin domains but shares similar flanking six-cysteine repeat thalmol. 12:450–454.
motifs. J. Biol. Chem. 275:39600–39607. 50. Schantz, P. M., D. Meyer, and L. T. Glickman. 1979. Clinical, serologic, and
30. Macchioni, G. 1999. A new species. Toxocara lyncis, in the caracal (Lynx epidemiologic characteristics of ocular toxocariasis. Am. J. Trop. Med. Hyg.
caracal). Parasitology 41:529–532. 28:24–28.
31. Magnaval, J. F., L. T. Glickman, P. Dorchies, and B. Morassin. 2001. 51. Schantz, P. M. 1989. Toxocara larva migrans now. Am. J. Trop. Med. Hyg.
Highlights of human toxocariasis. Korean J. Parasitol. 39:1–11. 41(Suppl.):21–34.
32. Magnaval, J. F., V. Galindo, L. T. Glickman, and M. Clanet. 1997. Human 52. Sharghi, N., P. Schantz, and P. J. Hotez. 2001. Toxocariasis: an occult cause
Toxocara infection of the central nervous system and neurological disorders: of childhood asthma, seizures, and neuropsychological deficit? Semin. Pedi-
a case-control study. Parasitology 115:537–543. atr. Infect. Dis. 32:E111–E116.
33. Maizels, R. M., K. K. A. Tetteh, and A. Loukas. 2000. Toxocara canis: genes 53. Shetty, A. K., and D. H. Aviles. 1999. Nephrotic syndrome associated with
expressed by the arrested infective larval stage of a parasitic nematode. Int. Toxocara canis infection. Ann. Pediatr. 19:297–300.
J. Parasitol. 30:495–508.
54. Small, K. W., B. W. McCuen, E. De Juan, and R. Machemer. 1989. Surgical
34. Maizels, R. M., and M. J. Holland. 1998. Parasite immunology: pathways for
management of retinal retraction caused by toxocariasis. Am. J. Ophthalmol.
expelling intestinal helminths. Curr. Biol. 8:R711–R714.
108:10–14.
35. Marmor, M., L. Glickman, and F. Shofer. 1987. Toxocara canis infection of
55. Sturchler, D., P. Schubarth, and M. Gualzata. 1989. Thiabendazole v. al-
children: epidemiologic and neuropsychologic findings. Am. J. Public Health
bendazole in treatment of toxocariasis: a clinical trial. Ann. Trop. Med.
77:554–559.
Parasitol. 83:473–478.
36. Mizgajska, H. 2001. Eggs of Toxocara spp. in the environment and their
56. Taylor, M. R., C. T. Keane, and P. O’Connor. 1988. The expanded spectrum
public health implications. J. Helminthol. 75:147–151.
of toxocaral disease. Lancet i:692–694.
37. Monshizadeh, R., M. T. Ashrafzadeh, and S. Rumelt. 2000. Choroidal neo-
vascular membrane: a late complication of inactive toxocara choroiretinitis. 57. Tariq, S. M., S. Matthews, and M. Stevens. 1997. Epidemiology of allergic
Retina 20:219–220. disorders in early childhood. Pediatr. Pulmonol. Suppl. 16:69.
38. Nathwani, D., R. B. Laing, and P. F. Currie. 1992. Covert toxocariasis as a 58. Reference deleted.
cause of recurrent abdominal pain in childhood. Br. J. Clin. Pract. 46:271. 59. Tost, F., A. Hellman, and G. Ockert. 1998. Toxocara canis infection. Envi-
39. Obwaller, A., E. Jensen-Jarolin. 1998. Toxocara infestations in humans: ronmental, parasitologic and epidemiologic studies. Ophthalmology 95:486–
symptomatic course of toxocariasis correlates significantly with levels of 489.
IgE/anti-IgE immune complexes. Parasite Immunol. 20:311–317. 60. Uga, S., and N. Kataoka. 1995. Measures to control Toxocara egg contam-
40. Obwaller, A., M. Duchene, and H. Bruhn. 2001. Recombinant dissection of ination in sandpits of public parks. Am. J. Trop. Med. Hyg. 52:21–24.
myosin heavy chain of Toxocara canis shows strong clustering of antigenic 61. Wilder, H. C. 1950. Nematode endophthalmitis. Trans. Am. Acad. Ophthal-
regions. Parasitol. Res. 87:383–389. mol. Otolaryngol. 55:99–104.
41. Oge, S., and H. Oge. 2000. Prevalence of Toxocara spp. eggs in the soil of 62. Wolach, B., Z. Sinnreich, and Y. Uziel. 1995. Toxocariasis: a diagnostic
public parks in Ankara, Turkey. Dtsch. Tier Wochenschr. 107:72–75. dilemma. Isr. J. Med. Sci. 31:689–692.
42. Oteifa, N. M., M. A. Moustafa, and B. M. Elgozamy. 1998. Toxocariasis as a 63. Worley, G., I. A. Green, and T. E. Frothingham. 1984. Toxocara canis infec-
possible cause of allergic diseases in children. J. Egypt Soc. Parasitol. 28: tion: clinical and epidemiological associations with seropositivity in kinder-
365–372. garten children. J. Infect. Dis. 149:591–597.
43. Page, A. P., W. Rudin, E. Fluri, M. L. Blaxter, and R. M. Maizels. 1992. 64. Xia, Y., Spence, H. J., and J. Moore. 2000. The ABA-1 allergen of Ascaris
Toxocara canis: a labile antigenic coat overlying the epicuticle of infective lumbricoides: sequence polymorphism, tissue-specific expression, lipid bind-
larvae. Exp. Parasitol. 75:72–86. ing function, and protein biophysical properties. Parasitology 120:211–224.
44. Prunier, F., S. Delpine, and J. Victor. 2001. Lofflerís fibroblastic endocardi- 65. Yahiro, S., G. Cain, and J. E. Butler. 1998. Identification, characterization,
tis. A report of a case complicating toxocariasis. Arch. Maladies Cour Vais- and expression of Toxocara canis nematode polyprotein allergen TBA-1.
seau 94:226–230. Parasite Immunol. 20:351–357.

You might also like