You are on page 1of 6

Review TRENDS in Parasitology Vol.22 No.

12

The future of Chagas disease control


Chris J. Schofield1, Jean Jannin2 and Roberto Salvatella3
1
London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
2
Neglected Tropical Diseases Department, World Health Organization (WHO), 1211 Geneva 27, Switzerland
3
Pan American Health Organization (PAHO), Montevideo CP 11300, Uruguay

In the past 15 years, there have been major advances in price – sometimes known as ‘el castigo del éxito’ (lit.
the control of Chagas disease in most of the countries punishment of success) – whereby success in reducing the
endemic for this infection. Attention now turns to epidemiological burden invariably also reduces political
the future continuity of surveillance and control interven- interest and operational budgets.
tions – especially in regions where control has been so Yet it is premature to believe that Chagas disease is
successful that the epidemiological significance of Cha- conquered. Experience from the programmes and cam-
gas disease is in steep decline. The effort and expenditure paigns against Triatominae throughout the Americas
of the recent past cannot continue indefinitely, but a demonstrates that all domestic populations of this subfam-
degree of surveillance and selective intervention will be ily can and should be eliminated. It is less clear whether
required because of the risk of new infestations and peridomestic populations of Triatominae can be so readily
infections resulting from adventitious silvatic vectors eliminated [1] and whether interventions should be contem-
accidentally entering houses. In this review, we summar- plated against silvatic populations [2]. Because available
ize the progress of multinational control initiatives techniques sometimes fail to eliminate all bugs inhabiting
against Chagas disease. In addition, we suggest that peridomestic habitats and because silvatic populations of
the most sustainable approach to future surveillance Triatominae are widespread in the Americas, even the
involves both the primary healthcare system and univer- successful elimination of domestic populations might not
sity-based teams, with progressively greater attention be sustained against occasional reinfestation from perido-
given to case detection and treatment. Such an idea is mestic or silvatic habitats. Even without re-establishment of
not new, but we believe that it merits extensive discus- the main domestic vectors such as T. infestans and Rhodnius
sion because of the different ways that research and prolixus, a low level of accidental transmission can be
health interventions are financed and because of the need expected because of silvatic or peridomestic species entering
to establish clearer reporting links between the research houses. The resulting transmission risks might be substan-
communities and the national health authorities. tially lower than those attributed to the original domestic
vectors – as shown by comparing human prevalence in areas
Is Chagas disease conquered? with Triatoma dimidiata, which was almost four times
On 9 June 2006, at their 15th annual meeting, the lower than in areas with R. prolixus [3] – but are not
Intergovernment Commission of the Southern Cone Initia- insignificant. Moreover, when an adventitious silvatic tria-
tive against Chagas disease formally declared Brazil to be tomine contaminates comestibles in a house, the consequent
free of Chagas disease transmission due to Triatoma infes- oral transmission can result in a localized ‘microepidemic’ of
tans. This represents a remarkable achievement considering acute cases of Chagas disease – as has occurred frequently in
that this species had been the primary domestic vector the Amazon region and elsewhere in Brazil [4–6]. In such
infesting rural houses in >700 municipalities of the 12 most situations, in which transmission occurs without the estab-
populated states of Brazil, in addition to vast areas of lishment of domestic vector populations, vector control
neighbouring countries of the Southern Cone (Figure 1). becomes of questionable priority compared with prompt
Throughout Latin America, the transmission of Trypano- diagnosis and specific treatment.
soma cruzi, the causative agent of Chagas disease (American The future scenario seems likely to involve a diverse
trypanosomiasis), has been steadily reduced through a ser- range of peridomestic and silvatic populations of Triato-
ies of multinational initiatives coordinated by the Pan Amer- minae, some of which might colonize houses and some of
ican Health Organization (PAHO: http://www.paho.org). In which might mediate the transmission of T. cruzi without
addition to Brazil, transmission has been effectively elimi- establishing domestic colonies. Such risks cannot be
nated in Uruguay (1997), Chile (1999), substantial areas of ignored and will require adequate approaches to surveil-
Argentina, Bolivia and Paraguay, and parts of Central lance, selective vector control, and treatment of new
America. Global disease prevalence has been reduced from human infections. However, this must be organized within
the 1990 estimates of 16–18 million people infected to the anticipated framework of reduced political interest and
9 million (Box 1) and can be expected to decline further reduced operational budgets.
through demographic changes and as a result of current
control interventions. But the health advances come at a
The balance of success
Corresponding author: Schofield, C.J. (cj.schofield@lshtm.ac.uk). The Southern Cone Initiative against Chagas disease
Available online 16 October 2006. followed a resolution by the Ministers of Health of
www.sciencedirect.com 1471-4922/$ – see front matter ß 2006 Elsevier Ltd. All rights reserved. doi:10.1016/j.pt.2006.09.011
584 Review TRENDS in Parasitology Vol.22 No.12

Box 1. The rise and fall of Chagas disease


Carlos Chagas first published his report of ‘a new human
trypanosomiasis’ in 1909 [39]. In the following years, there was
extensive argument about whether the discovery was important, or
even real [40,41], with the unnecessary polemic continuing even to
the end of the 20th century [42]. The arguments were trivial, at times
scurrilous, but probably contributed to the successive negation of a
well-deserved Nobel Laureate in recognition of Chagas’ outstanding
and detailed studies [41,43]*.
By 1960, Chagas disease had been reported from all countries of
Latin America, and the first WHO Expert Committee meeting on
Chagas disease estimated global prevalence of the infection to be 7
million people, with perhaps another 35 million at risk [44].
Successive reports revised these estimates steadily upwards to a
peak of 24 million people thought to be infected in the mid-1980s
[45]. By the end of the 1980s, data from wide-ranging serological
surveys provided more detailed estimates for most countries,
leading to the oft-quoted figures of 16–18 million people infected
with Chagas disease, with a further 90–100 million at risk [46,47].
Figure 1. Apparent distribution of Triatoma infestans. (a) The maximum predicted From the same dataset of seroprevalence, annual incidence could
distribution (in the absence of control interventions) from a geographic be calculated at 450 000 new infections per year in the absence of
information system (GIS) thematic analysis, which reveals a few points outside control interventions, and the World Bank (http://www.worldbank.
the Southern Cone region (and in southern Chile and around the Peru–Bolivia– com) ranked Chagas disease as the most important parasitic disease
Brazil border) that might be suitable for T. infestans but from where this species
of the Americas in terms of its socioeconomic impact, estimated as
has never been recorded. The predicted maximum distribution of T. infestans
covers 6 278 081 km2 [38]. (b) An estimate of current distribution based on reports
disability-adjusted life years (DALYs) lost because of the infection
from the Intergovernment Commission for the Southern Cone Initiative, from [48].
which the estimated distribution of T. infestans has been reduced to 913 485 km2. With the success of the Southern Cone Initiative, launched in
Part (a) provided by David E. Gorla. 1991, and similar initiatives launched in 1997 for Central America
and the Andean Pact countries, prevalence estimates have been
steadily declining. As early as 1994, from reports of reduced
Argentina, Bolivia, Brazil, Chile, Paraguay and Uruguay, incidence in Argentina and Brazil, overall prevalence was estimated
meeting in Brasilia (Brazil) in 1991. It focused on the to be 14 615 000 [49]. Estimates for these two countries, and Chile
interruption of T. cruzi transmission by eliminating and Venezuela were then further reduced, although prevalence
domestic vectors (particularly T. infestans), together with estimates for Ecuador and Mexico were revised upwards as a result
of more-extensive serological surveys, leading to an overall
extended screening of blood donors to reduce the risk of
estimated prevalence of 11 253 375 in 1998 [50]. The most recent
transfusional transmission, and the promotion of maternal estimate, from the Disease Control Priorities Project of the NIH
screening for infection followed by specific treatment of (http://www.nih.gov) and World Bank, indicates an overall preva-
infected newborns. Similar initiatives were agreed in 1997 lence of 9.8 million people [51].
*
for the Central American and Andean Pact countries, and Carlos Chagas was formally nominated twice for the Nobel Prize in Medicine,
in 1913 and 1921.
in 2004 a surveillance initiative was announced for the
nine countries of the Amazon basin.
The progress of these initiatives has been widely
reviewed [7–12]. Extensive screening of blood donors for chronically infected mothers seems to be declining in areas
T. cruzi infection is now carried out in most countries of the where vectorial transmission has been interrupted.
Americas (Table 1), and the distribution of domestic The Chagas disease control interventions also seem to
vectors has been markedly reduced, with transmission have been extremely beneficial in financial terms. In
interrupted over vast areas. Because the rate of new Brazil, for example, annual treatment costs for Chagas
infection has declined to zero over such substantial areas, disease were estimated to be approximately US$200
estimates of prevalence have been progressively reduced to million per year in 1996, compared with the annual invest-
a current figure of 9.8 million people infected (Box 1). Of the ment in control of approximately US$20 million; in Chile,
100 million people thought to be at risk from Chagas annual treatment costs were estimated to be US$14–19
disease in 1990, 60 million now sleep without the risk million, compared with annual intervention costs of
of infection and without the nuisance and chronic blood loss approximately US$300 000. For the Southern Cone
previously caused by domestic colonies of Triatominae. countries as a whole, aggregate costs since 1991 have been
The medical benefits of the control initiatives are approximately US$320 million [9], which is within the
reflected in the progressive decline of hospitalizations initial estimates of US$190–350 million [14]. The internal
for Chagas disease [9]. This is primarily because of the rate of return on this investment was initially predicted at
reduced numbers of new acute infections but also seems to just over 14% [11] but point studies show actual rates of
reflect a declining severity of chronic disease. This decline return of 30% in Brazil [15] and >60% for the province of
in severity might be a result of halting reinfection, an idea Salta in Argentina [16].
recently endorsed by studies of chronically infected mice in But, against the social, medical and financial successes
which rates of severe cardiac lesions were significantly of the Chagas disease control initiatives, some difficulties
lower in mice infected only once compared with mice that must be acknowledged. Although 60 million people no
were repeatedly reinfected [13]. A similar effect might longer share their homes with blood-sucking Triatominae,
be reducing the likelihood of congenital infection because 40 million people remain at risk. Although 5 million
the apparent rate of transplacental transmission from new infections of Chagas disease have probably been
www.sciencedirect.com
Review TRENDS in Parasitology Vol.22 No.12 585

Table 1. Screening of blood donors for Trypanosoma cruzi in R. prolixus from Central America, which seems to have
Latin Americaa been accidentally imported from a Venezuelan origin early
Coverage (%) Seropositive (%) in the 20th century [23,24]. Such arguments might also be
Southern Cone countries
applicable to populations of T. dimidiata in Ecuador and
Argentina 100 4.50
Bolivia 86 9.90 northern Peru, which seem to have been accidentally
Brazil 100 0.61 imported in pre-Columbian times [25,26], and to Rhodnius
Chile 75 b 0.47 ecuadoriensis in parts of northern Peru, which might
Paraguay 99 2.80 have been imported within the past 50 years [27]. To a
Uruguay 100 0.47
large extent, however, much of what seemed possible in
Andean Pact countries
Colombia 99 0.98
these elimination concepts has already been achieved. T.
Ecuador 100 0.15 infestans does seem to have been eliminated over much of
Peru 99 0.26 its range, R. prolixus is disappearing from Central Amer-
Venezuela 100 0.67 ica, and the recently launched national programme in
Central America Ecuador will probably have substantial success against
Belize 100 0.40
100 0.34
domestic T. dimidiata.
Costa Rica
El Salvador 100 2.46 The imperative to eliminate all domestic populations of
Guatemala 100 0.79 Triatominae derives not only from their capacity to trans-
Honduras 100 1.40 mit T. cruzi but also from their high nuisance value and
Nicaragua 100 0.49 contribution to chronic blood loss [28,29]. Although this
Panama 98 0.90
a
must remain central to future programmes, the underlying
Table based on data from Ref. [26], with revisions from 2006 Intergovernment
Commission reports.
rationale will be different. The idea of large-scale elimina-
b
98% in endemic regions. tion was justified as a strategy to prevent future reinfesta-
tions by that species. However, where reinfestation is
expected to be less frequent and probably due to local
prevented, transmission is still apparent in several coun- silvatic species, the more appropriate strategy will be of
tries and might even be increasing in parts of Argentina periodic intervention when householders report new infes-
[17], Venezuela [18] and the Amazon region [4]. Mexico, tations. The effectiveness of such an idea depends on three
Peru, Colombia and Costa Rica still have no national features: (i) a system to receive and acknowledge house-
programme for the control of Chagas disease vectors, holder reports; (ii) a system to collate such reports with
and programmes for screening blood donors remain below other information of operational and epidemiological rele-
targets in Bolivia and Mexico. But perhaps the greatest vance; and (iii) capacity to implement selective interven-
difficulty lies in the premature idea that Chagas disease tions in response to reports of individual and/or clustered
has been conquered, and the consequent dismemberment infestations. Such a design has already been proposed and
of executive services responsible for surveillance and con- tested for the control of Chagas disease in areas of Central
trol. Today, only four countries in the Americas – Nicar- America where the main vector is T. dimidiata (Box 2).
agua, Ecuador, Paraguay and Panama – retain an This species retains silvatic and peridomestic populations
executive structure with national responsibility for the in many areas, from which it can gradually recolonize
control of Chagas and other vector-borne diseases. In the houses from which the original domestic population has
other countries, responsibility has been devolved to state, been eliminated. The proposed strategy involves an initial
provincial and municipal authorities, with – at best – a programme to eliminate all existing domestic populations,
centralized authority retained only for statistics and leg- followed by an annual cycle of accumulating reinfestation
islation. Such decentralization risks heterogeneity of reports and an annual round of selective interventions in
action (and inaction), discontinuity of programme imple- response. In parallel, progressively greater emphasis is
mentation (especially of surveillance) and inefficiencies given to active case detection and treatment, especially
due to duplication of administrative effort – precisely among school-age children.
the problems that the multinational initiatives, with all
countries working simultaneously within a coordinated Case detection and treatment
framework, were designed to avoid. The change from an elimination strategy to one of
continual selective intervention carries implicit acceptance
Vector control strategy that occasional accidental transmission might occur –
When the Southern Cone programme was launched in chiefly due to adventitious silvatic vectors entering houses.
1991, it was believed that the main vector in the countries In the past, such an idea was seen to be problematic
affected, T. infestans, could be eliminated over most of its because of the belief that treatment of human cases would
range. This idea was based on historical reconstruction [19] be satisfactory only if administered within the early acute
progressively supported by genetic studies [20–22] indicat- phase of infection, although accumulating experience
ing that, in most Southern Cone countries, T. infestans had begins to indicate otherwise.
been accidentally imported in relatively recent times, par- The two drugs available for specific treatment of T.
ticularly during the past 100 years. The control interven- cruzi infection are nifurtimox (Lampit1), launched by
tions could be seen as correcting a historical accident and Bayer (http://www.bayer.com) in 1967, and benznidazole
restoring the vector distribution to its origins. Similar (Rochagan1, Radanil1), launched by Roche (http://
arguments were implicit in the idea of eliminating www.roche.com) in 1972. Initially shown to be effective
www.sciencedirect.com
586 Review TRENDS in Parasitology Vol.22 No.12

the feasibility and the most appropriate techniques for


Box 2. Steps in the proposed operational strategy for the
active case detection of asymptomatic Chagas disease; this
control of Triatoma dimidiata in Central America
is actively pursued in Central America, using the Chagas
The following information was adapted from Ref. [52]. Stat-Pak1 for a rapid assessment of schoolchildren [36],
1. In each school in the endemic areas, serological testing of a followed by quantitative ELISA to confirm positivity and
random sample of 30 schoolchildren using the Chagas
Stat-Pak1 and a questionnaire for possible recognition of
monitor changes in serological titres after treatment (Box
Triatominae. 2). In some countries, however, treatment is discouraged
(a) If all tests are negative and no child reports having seen T. for non-acute cases, and may be authorized only after
dimidiata in their house, the school catchment area apparently residual house spraying to remove vectors.
has no active transmission.
(b) If three or fewer children are positive for Chagas disease, the
school catchment area is considered a medium priority for
Outline for the future
entomological survey and control interventions. All positive The future scenario for Chagas disease control must
children given quantitative ELISA for confirmation, followed by continue to be based on the idea of eliminating all existing
specific treatment with benznidazole or nifurtimox. domestic populations of Triatominae but then accepting
(c) If more than three children are positive, the school catchment
that reinfestation might occur or that adventitious silvatic
area is considered a high priority for entomological survey and
control interventions. All children in school are tested by Chagas
Triatominae might enter a house and cause disease trans-
Stat-Pak1, with all positives given quantitative ELISA for mission without establishing a new domestic colony – as in
confirmation, followed by specific treatment with benznidazole Amazonia. In such a scenario, large-scale vector control
or nifurtimox. campaigns of the style of the multinational initiatives
2. School catchment areas can now be ranked in terms of
would become progressively less relevant as existing
apparently active transmission. In this order, they can be visited
for entomological surveys in each house, followed by residual domestic infestations are eliminated. However, surveil-
spraying of all houses confirmed to be infested; all houses are lance and focal interventions against newly established
identified by their geographical coordinates, and all house- domestic colonies of Triatominae would be required. In
holders are informed about T. dimidiata and asked to report all addition, improved parasitological surveillance would be
such insects to the local health centre.
3. Health centre reports are collated and mapped during the year.
warranted, with the idea of offering specific treatment to
Isolated reports of infestation can be checked, with the houses all new cases of infection and probably to asymptomatic
sprayed if infestation is confirmed; if several reports indicate a chronic cases. The key issue is how to maintain entomo-
cluster of infested houses, all houses in the apparent cluster can logical and parasitological surveillance, especially as the
be checked and sprayed. incidence of new infestation and infection declines. Much is
4. If, during a reporting year, no reports are received from houses
in what was a previous infestation cluster, the cluster can be
written about the importance of community participation,
checked to make sure that the absence of reports is not due to a and it is indeed a key component both of the initial inter-
breakdown of the reporting system. ventions and of the subsequent surveillance, but commu-
5. Steps 1 and 2 can be repeated in accordance with epidemio- nity interest wanes when there is little to report, so an
logical assessment and available resources.
additional approach is required. This could be provided by
university-based research teams [37] but would be of little
at curing acute infections, these drugs were largely relevance without a clear reporting structure so that data
discarded for treatment of chronic T. cruzi infections can be collated and selective interventions undertaken
because of a high likelihood of serious side-effects com- where necessary.
bined with an apparently low likelihood of radical cure Perhaps one of the greatest achievements of the
[30]. Such ideas are being challenged by evidence that multinational initiatives against Chagas disease is that
serious side-effects are much less frequent in younger surveillance and control of the disease and its vectors are
age-groups and that reversion to seronegativity after currently on the agenda of all endemic countries, even
treatment can occur in a large proportion of patients – those such as Belize, Guyana and Suriname, where the
especially children – although this might not occur for problem has been minimal. This means that a degree of
several years [31,32]. National policy in several countries vector and disease surveillance will be carried out in all of
is now to offer specific treatment to all cases that occur in these countries so that, with adequate data collation, it
patients under 14–15 years of age, whether acute or should be possible to detect epidemiological trends and
chronic, and current research on adults in Argentina potential new outbreaks of transmission. But such an idea
indicates that this policy could be extended to a much must emphasize the need for adequate collation of avail-
wider range of patients [33–35]. able data at national and regional levels, which at present
The World Health Organization (WHO: http:// is often lost between official reports and formal publication.
www.who.int) is working in partnership with industry to There is currently no international system – beyond review
ensure adequate supplies of both drugs. Nifurtimox is now articles – that can monitor and collate epidemiological data
manufactured by Bayer in El Salvador and can be made over a continental scale.
available free of charge through WHO and PAHO; the
supply of benznidazole has been guaranteed by Roche until Concluding remarks: towards a sustainable end-point
adequate production can be sustained by the Brazilian The most consistently successful programme against
laboratory Laboratório Farmacêutico do Estado de Chagas disease has been that of the Brazilian state of
Pernambuco (LAFEPE) – although neither the cost of Sao Paulo – a programme initiated with a state-wide ‘attack
the drug nor procedures for its distribution outside Brazil phase’ in 1964–1967 and continued through a series of
have been finalized. A further issue to be resolved concerns strategic adjustments in accordance with the changing
www.sciencedirect.com
Review TRENDS in Parasitology Vol.22 No.12 587

At national and regional levels, however, the focus in


Box 3. Current and future phases in Chagas disease control
recent decades has been on eliminating the primary
Phase 1 domestic vectors. Less thought has been given to the design
1.1 Elimination of all existing domestic populations of Triatominae; of an institutionally stable end-point to maintain the
this requires one or two spray rounds, designed to treat all
houses in infested localities in the case of Triatoma infestans or
advances, prevent recrudescence of transmission and
Rhodnius prolixus, or targeting only those confirmed to be respond adequately to new cases that might occur. Such
infested with other vectors. a system will depend crucially on a capacity to seek and
1.2 Organization of community-based surveillance networks. These assimilate georeferenced epidemiological data – from
report the presence of residual (or new) domestic infestations to whatever source – and to organize serological screening,
a local voluntary post (posto de informacion sobre triatomi-
neos), with adequate reporting to the local health authorities,
and treatment of new cases, together with selective vector
entomological confirmation and written acknowledgement of control interventions where necessary. Medical profes-
householder reports, followed by selective intervention where sionals and the primary healthcare system, together with
required or requested. schools (Box 2), would have a fundamental role but only
1.3 Improved and standardized screening of blood donors (with
with adequate training and clear reporting lines to the
clinical follow-up and psychological counselling for those found
to be infected). epidemiological surveillance centre. In many countries,
Phase 2 academic research teams also have a significant role in
2.1 Continuation and support for community-based surveillance the surveillance and control of Chagas disease, and this
networks, with selective interventions carried out by local health
should be encouraged as an important complement to the
authorities.
2.2 Development of the community networks as general health health surveillance structure. It is a way to accord national
surveillance systems linked to the primary healthcare system of relevance to research, improve interactions between
that locality. health and education authorities, and might come to
2.3 Routine blood-slide microscopy from all febrile cases (com- represent the primary source of specific epidemiological
bined with haemoconcentration techniques where possible) to
information.
diagnose new infections and offer prompt specific treatment as
necessary.
2.4 Encouragement of university-based field teams to carry out Acknowledgements
investigative projects designed to evaluate the progress of This review has benefited from international collaboration through the
Chagas disease control and study the biology of non-dom- Latin American Network for Research on the Biology and Control of
iciliated vectors in specific regions (this could involve logistic Triatominae (ECLAT). We also thank Sergio Sosa-Estani for additional
support from the local health authorities and financial support information about current trials of specific treatment for Chagas disease
from the national research councils). in chronically infected adults, and David E. Gorla for access to
Phase 3 unpublished geographic information systems (GIS) studies of Triatoma
3.1 Development of a national database for epidemiological infestans distribution.
surveillance of Chagas disease using GIS techniques, with
mandatory reporting of all new cases of infection and all
findings of domestic, peridomestic and silvatic populations of
References
1 Cecere, M.C. et al. (2006) Comparative trial of the effectiveness of
Triatominae (such reporting could be a condition of research
pyrethroid insecticides against peridomestic populations of Triatoma
council support for academic research teams and of national
infestans in northwestern Argentina. J. Med. Entomol. 43, 902–909
support to local health centres).
2 Guhl, F. and Schofield, C.J., eds (2004) Proceedings of the
3.2 Development of the Chagas database and reporting system for
ECLAT–AMCHA International Workshop on Chagas disease
other pathologies, ideally linked to a small ‘rapid deployment
surveillance in the Amazon Region, Palmari, Brasil, Universidad de
force’ of highly trained professionals able to support emergency
Los Andes
vector control or public health interventions in all parts of the
3 Paz Bailey, G. et al. (2002) Incidence of Trypanosoma cruzi infection
country.
in two Guatemalan communities. Trans. R. Soc. Trop. Med. Hyg. 96,
Phase 1 is well advanced in most countries, and a small number –
48–52
notably Brazil and some Central American countries – is advancing
4 Coura, J.R. et al. (2002) Emerging Chagas disease in Amazonian
with phase 2. None, however, has yet advanced to phase 3.
Brazil. Trends Parasitol. 18, 171–176
5 Steindel, M. et al. (2005) Doença de Chagas: mal que ainda preocupa.
Ciencia Hoje 37, 32–38
epidemiological circumstances (D. Wanderley, PhD thesis, 6 Benchimol Barbosa, P.R. The oral transmission of Chagas disease: an
Universidade de Sao Paulo, Brazil, 1994). Although acute form of infection responsible for regional outbreaks. Int. J.
T. infestans was probably eliminated from Sao Paulo by Cardiol. (in press)
1990, a system of community-based surveillance is main- 7 Dias, J.C.P. and Schofield, C.J. (1999) The evolution of Chagas disease
(American trypanosomiasis) control after 90 years since Carlos
tained for secondary vectors (e.g. Triatoma sordida and Chagas discovery. Memórias do Inst. Oswaldo Cruz 94 (Suppl. 1),
Panstrongylus megistus), together with centralized data 103–121
assimilation and selective interventions where necessary. 8 Dias, J.C.P. and Schofield, C.J. (2004) Control of Triatominae. In The
Sao Paulo has been effectively free of Chagas disease trans- Trypanosomiases (Maudlin, I. et al., eds), pp. 547–563, CAB
mission since the early 1990s and offers a model for the International
9 Dias, J.C.P. et al. (2002) The impact of Chagas disease control in Latin
various phases in Chagas disease control. This includes the America. Memórias do Inst. Oswaldo Cruz 97, 603–612
essential idea that, although domestic vector populations 10 Schofield, C.J. and Dias, J.C.P. (1999) The Southern Cone Initiative
can and should be eliminated, there will be a perennial need against Chagas disease. Adv. Parasitol. 42, 1–27
for surveillance, data assimilation and selective interven- 11 Yamagata, Y. and Nakagawa, J. (2006) Control of Chagas disease. Adv.
Parasitol. 61, 129–165
tions. In addition, a primary healthcare approach is
12 Schmunis, G.A. and Cruz, J.R. (2005) Safety of the blood supply in
required to detect and treat new cases of Chagas disease Latin America. Clin. Microbiol. Rev. 18, 12–29
(Box 3) – although this will require a consensus about the 13 Bustamante, J.M. et al. (2002) Trypanosoma cruzi reinfections in mice
most appropriate diagnostics and treatment regime. determine the severity of cardiac damage. Int. J. Parasitol. 32, 889–896
www.sciencedirect.com
588 Review TRENDS in Parasitology Vol.22 No.12

14 Schofield, C.J. and Dias, J.C.P. (1991) A cost–benefit analysis of 34 Sosa-Estani, S. (2005) Red Latinoamericana de investigadores.
Chagas disease control. Memórias do Inst. Oswaldo Cruz 86, 285–295 Evaluación de la eficacia del tratamiento con benznidazol en
15 Akhavan, D. (2000) Análise de Custo-Efetividade do Programa de pacientes adultos con enfermedad de Chagas crónica. Proyecto
Controle da Doença de Chagas no Brasil, Organização Pan-Americana BENEFIT. Sitio Argentina. In VII Congreso Argentino de
da Saúde Protozoologı́a y Enfermedades Parasitarias, Summary Book MR 5,
16 Basombrio, M.A. (1998) A cost–benefit analysis of Chagas disease pp. 29–30, Mendoza, Argentina
control in northwest Argentina. Trans. R. Soc. Trop. Med. Hyg. 92, 35 Viotti, R. et al. (2006) Long-term cardiac outcomes of treating chronic
137–143 Chagas disease with benznidazole versus no treatment: a
17 Gurtler, R.E. (2005) Incidence of Trypanosoma cruzi infection among nonrandomized trial. Ann. Intern. Med. 144, 724–734
children following domestic reinfestation after insecticide spraying in 36 Ponce, C. et al. (2005) Validation of a rapid and reliable test for
rural northwestern Argentina. Am. J. Trop. Med. Hyg. 73, 95–103 diagnosis of Chagas disease by detection of Trypanosoma cruzi-
18 Feliciangeli, M.D. et al. (2003) Chagas disease control in Venezuela: specific antibodies in blood of donors and patients in Central
lessons for the Andean region and beyond. Trends Parasitol. 19, 44–49 America. J. Clin. Microbiol. 43, 5065–5068
19 Schofield, C.J. (1988) The biosystematics of Triatominae. In 37 Dujardin, J.P. and Schofield, C.J. Vector control by surveillance
Biosystematics of Haematophagous Insects (Systematics Association networks: the ECLAT programme and Chagas. In Encyclopedia
Special Volume 37) (Service, M.W., ed.), pp. 284–312, Oxford of Infectious Diseases: Modern Methodologies, Wiley & Sons
University Press (in press)
20 Dujardin, J.P. et al. (1998) Population structure of Andean Triatoma 38 Gorla, D.E. (2002) Variables ambientales registradas por sensores
infestans: allozyme frequencies and their epidemiological relevance. remotos como indicadores de la distribución geográfica de Triatoma
Med. Vet. Entomol. 12, 20–29 infestans. Ecol. Aust. 12, 117–127
21 Panzera, F. (2004) Genomic changes of Chagas disease vector, South 39 Chagas, C. (1909) Nova trypanosomiase humana. Gaceta Medica da
America. Emerg. Infect. Dis. 10, 438–446 Bahia 40, 433–440
22 Bargues, M.D. et al. (2006) Origin and phylogeography of the Chagas 40 Lewinsohn, R. (1979) Carlos Chagas (1879–1934): the discovery of
disease main vector Triatoma infestans based on nuclear rDNA Trypanosoma cruzi and of American trypanosomiasis (footnotes to
sequences and genome size. Infect. Genet. Evol. 6, 46–62 the history of Chagas disease). Trans. R. Soc. Trop. Med. Hyg. 73,
23 Dujardin, J.P. (1998) The origin of Rhodnius prolixus in Central 513–523
America. Med. Vet. Entomol. 12, 113–115 41 Lewinsohn, R. (2003) Três Epidemias Lições do Passado, Editora
24 Zeledón, R. (2004) Some historical facts and recent issues related to the Unicamp
presence of Rhodnius prolixus (Stal,1859) (Hemiptera,Reduviidae) in 42 Delaporte, F. (1999) La Maladie de Chagas, Bibliotheque Scientifique
Central America. Entomologia y Vectores 11, 233–246 Payot
25 Abad-Franch, F. (2001) Biogeography of Triatominae in Ecuador: 43 Coutinho, M. and Dias, J.C.P. (2000) The rise and fall of Chagas
implications for the design of control strategies. Memórias do Inst. disease. Perspect. Sci. 7, 447–485
Oswaldo Cruz 96, 611–620 44 WHO (1960) Chagas Disease. Report of a Study Group. (WHO Technical
26 Schofield, C.J. (2002) Evolución y control del Triatoma dimidiata. In Report Series 202), World Health Organization
Taller Para el Establecimiento de Pautas Tecnicas en el Control de 45 Walsh, J.A. (1984) Estimating the burden of illness in the tropics. In
Triatoma dimidiata, San Salvador, 11–13 March 2002. pp. 12–18, Tropical and Geographical Medicine (Warren, K.S. and Mahmoud,
PAHO document OPS/HCP/HCT/214/02, Organización Panamericana A.A.F., eds), pp. 1073–1085, McGraw-Hill
de la Salud 46 WHO (1990) Tropical Diseases 1990. TDR/CTD/HH20.1, World Health
27 Schofield, C.J. and Dujardin, J.P. (1999) Theories on the evolution of Organization
Rhodnius. Actualidades Biologicas 70, 183–197 47 WHO (1991) Control of Chagas Disease. (WHO Technical Report Series
28 Rabinovich, J.E. (1979) Domiciliary biting frequency and blood 811), World Health Organization
digestion of the Chagas disease vector Rhodnius prolixus Stahl 48 World Bank (1993) World Development Report 1993. Investing in
(Hemiptera: Reduviidae) in Venezuela. Trans. R. Soc. Trop. Med. Health, Oxford University Press
Hyg. 72, 272–283 49 OPS (1994) Las Condiciones de Salud de las Americas. (Publicacion
29 Schofield, C.J. (1981) Chagas disease, triatomine bugs, and blood-loss. Cientifica 549) (Vol. 1), pp. 147–149, Organização Pan-Americana
Lancet 1, 1316 da Saúde
30 Cancado, J.R. and Brener, Z. (1979) Terapêutica. In Trypanosoma cruzi 50 Schmunis, G.A. (1999) Proceedings of the Second International
e Doença de Chagas (Brener, Z. and Andrade, Z., eds), pp. 362–424, Workshop on Population Biology and Control of Triatominae,
Guanabara Koogan Tegucigalpa, Honduras: Initiativa del Cono Sur. pp. 26–31, INDRE
31 Pinto Dias, J.C. (2006) The treatment of Chagas disease (South 51 Remme, J.H.F. et al. (2006) Tropical diseases targeted for
American trypanosomiasis). Ann. Intern. Med. 144, 772–774 elimination: Chagas disease, lymphatic filariasis, onchocerciasis
32 Castro, A.M. et al. Detection of parasitemia profiles by blood culture and leprosy. In Disease Control Priorities in Developing Countries
after treatment of human chronic Trypanosoma cruzi infection. (2nd edn), pp. 433–449, World Bank/Oxford University
Parasitol. Res. (in press) Press
33 Sosa-Estani, S. and Segura, E.L. (1999) Treatment of Trypanosoma 52 Ponce, C. and Schofield, C.J. (2004) Strategic options for the control
cruzi infection in the indeterminate phase: experience and current of Triatoma dimidiata in Central America. In Iniciativa de los
guidelines in Argentina. Medicina (Buenos Aires) 59 (Suppl. 2), Paises Centroamericanas, Organización Panamericana de la
166–170 Salud

www.sciencedirect.com

You might also like