Chapter One - Chagas Disease Diagnostic Applications: Present Knowledge and Future Steps

https://doi.org/10.1016/bs.apar.2016.10.001Get rights and content

Abstract

Chagas disease, caused by the protozoan Trypanosoma cruzi, is a lifelong and debilitating illness of major significance throughout Latin America and an emergent threat to global public health. Being a neglected disease, the vast majority of Chagasic patients have limited access to proper diagnosis and treatment, and there is only a marginal investment into R&D for drug and vaccine development. In this context, identification of novel biomarkers able to transcend the current limits of diagnostic methods surfaces as a main priority in Chagas disease applied research. The expectation is that these novel biomarkers will provide reliable, reproducible and accurate results irrespective of the genetic background, infecting parasite strain, stage of disease, and clinical-associated features of Chagasic populations. In addition, they should be able to address other still unmet diagnostic needs, including early detection of congenital T. cruzi transmission, rapid assessment of treatment efficiency or failure, indication/prediction of disease progression and direct parasite typification in clinical samples. The lack of access of poor and neglected populations to essential diagnostics also stresses the necessity of developing new methods operational in point-of-care settings. In summary, emergent diagnostic tests integrating these novel and tailored tools should provide a significant impact on the effectiveness of current intervention schemes and on the clinical management of Chagasic patients. In this chapter, we discuss the present knowledge and possible future steps in Chagas disease diagnostic applications, as well as the opportunity provided by recent advances in high-throughput methods for biomarker discovery.

Introduction

Chagas disease or American Trypanosomiasis, caused by the parasitic protozoan Trypanosoma cruzi (Kinetoplastida, Trypanosomatidae), is a life-long, neglected tropical disease and leading cause of cardiomyopathy in endemic areas (Rassi et al., 2010). With 8–10 million people already infected and up to 120 million individuals at risk of infection, Chagas disease constitutes the most important parasitic disease in Latin America and one of the most common globally (Stanaway and Roth, 2015). Its exact burden is, however, difficult to assess due to several factors including the widespread geographic distribution of T. cruzi vectorborne transmission, the decades-long lag between infection and appearance of symptoms, certain pitfalls of current diagnostic methods, biased prevalence data and incomplete recognition of Chagas disease-attributable symptoms (Stanaway and Roth, 2015). The most recent estimates indicate that Chagas disease is responsible for ∼550,000 disability-adjusted life years (DALYs), a measure that captures both premature mortality (∼12,000 deaths per year) and nonfatal health losses (Stanaway and Roth, 2015). Despite this enormous toll, only two trypanocydal drugs, benznidazole and nifurtimox, are currently available for chemotherapy. Both are nitroheterocyclic, oral compounds that require prolonged administration, may display severe adverse effects, cannot be used to treat pregnant women due to their uncertain teratogenic risks and, most importantly, show high efficacy solely if administered at the onset of infection (Carlier and Truyens, 2015, Rassi et al., 2010, Viotti et al., 2006). The prospects for the development of an effective vaccine for prophylactic and/or therapeutic purposes, on the other hand, are still clouded by substantial scientific and socioeconomic challenges (Beaumier et al., 2016, Bustamante and Tarleton, 2015).

T. cruzi transmission primarily occurs when humans are exposed to the contaminated feces of infected, haematophagous triatomine vectors. Large-scale intervention schemes launched in different regions of Latin America in the 1990s have successfully shrunk the geographic limits and prevalence of vectorborne parasite transmission and led to an overall ∼40% reduction of disease prevalence (Schofield et al., 2006). However, different ecological and demographic issues converged in the last decades to shift the epidemiological landscape for this disease. For instance, recent outbreaks of acute cases in certain regions from Brazil and Venezuela were not strictly vectorborne but rather due to accidental ingestion of T. cruzi-tainted food and fluids (Alarcon de Noya et al., 2010, Segovia et al., 2013). This ‘foodborne’ transmission mode likely constitutes an ancient epidemiological trait, very important to the zoonotic spreading of the parasite (Gurtler and Cardinal, 2015), and appears to be associated with increased virulence and a higher case-fatality rate in humans (Alarcon de Noya et al., 2010, Segovia et al., 2013). In addition, migratory trends of infected populations from rural areas to urban centres and/or to nonendemic regions along with changes in the ecogeographical distribution of vector populations have led to the gradual urbanization and globalization of Chagas disease, which is now recognized as an emerging worldwide threat to public health (Eisenstein, 2016). Indeed, the risk of acquiring Chagas disease through infected blood transfusion and organ transplantation is becoming a major problem even in areas of nonendemicity, such as the United States, Australia and Europe (Requena-Mendez et al., 2015, Schmunis and Yadon, 2010). Moreover, the congenital route of infection, which constitutes the main transmission mode of T. cruzi in nonendemic areas, is now estimated to be responsible for 22% of new annual infections in endemic countries with active programs for home vector infestations control (Carlier and Truyens, 2015).

In this scenario, a strong and global partnership aimed to coordinate actions to control parasite transmission is urgently needed. In particular, we need to redouble our efforts to control home vector infestation, to screen blood supplies and to identify and subsequently treat T. cruzi-infected people who are still in the early stages of the disease to avoid sequelae, morbidity and economic losses. As a major step towards these goals, we ought to develop novel biomarkers able to overcome the limitations of current diagnostic applications. In this chapter, we critically appraise what has so far been achieved in this area. We also discuss possible ways to proceed to address major and still unmet diagnostic demands and the opportunity provided by recent advances in high-throughput methods (i.e., peptide synthesis technology, genomics and proteomics) in Chagas disease biomarker discovery.

Section snippets

Trypanosoma cruzi, an ‘all-wheel drive’ parasite

T. cruzi is a promiscuous parasite that traverses a complex life cycle involving extracellular proliferation and differentiation inside haematophagous insect vectors from different genera and intracellular proliferation and differentiation in a variety of vertebrate hosts (De Souza, 2002). Host switching, immune pressure as well as constant transition from intracellular to extracellular niches (and vice versa) pose significant adaptation challenges and are concomitantly accompanied by extensive

Parasitological and clinical methods

Upon T. cruzi infection, patients undergo the acute phase of Chagas disease, which extends for 40–60 days. Symptoms, if indeed occur, are usually very mild and atypical, thus often misleading its clinical recognition (Rassi et al., 2010). In rare cases of vectorborne transmission, a skin nodule (called ‘chagoma’) or painless prolonged eyelid oedema (called the ‘Romanha's sign’) may indicate the site of parasite inoculation. Due to the patent parasitaemia verified at this initial phase,

Diagnostic Applications for Chagas Disease: Pending Issues

As described earlier, current diagnostic methods and particularly those based on serology are highly accurate in detecting most of T. cruzi infections in humans. However, there are some clinical and/or epidemiological situations, discussed in this section, in which their performance is significantly hampered by methodological and/or biological issues.

Diagnostic Applications for Chagas Disease: The Road Ahead

As discussed throughout this chapter, parasite-specific immune signatures provided a prime source of biomarker candidates for development of Chagas disease diagnostic applications (see Table 1). However, they may now be reexplored using modern and powerful ‘omics’-based fingerprint approaches. Indeed, the availability of complete T. cruzi genomes from several strains together with a variety of recent genome mining exercises was performed to identify potential serodiagnostic reagents and vaccine

Concluding Remarks

Over 100 years after its discovery, and despite its huge medical, economic and social burden, Chagas disease remains a major threat in several countries of Latin America and an emergent global health problem. Great efforts have been made and are still being made in Latin America and other developed countries to halt T. cruzi transmission. However, one of the key issues concerning Chagas disease control remains that of diagnosis. Without accessible and effective diagnostics tools and methods,

Acknowledgement

We express our gratitude to Dr. Carlos Frasch (IIB-INTECh) for critical reading of the manuscript and to Dr. Javier Di Noia (IRCM, Canada) for the enthusiasm and superb graphic art. We apologize to people whose work was not referenced due to limited space. Research carried out in our labs is supported by grants and contracts from the Agencia Nacional de Promoción Científica y Tecnológica (ANPCyT, Argentina) (to FA and CAB), the National Institutes of Health (NIAID/NIH, USA) (to FA), and

References (261)

  • J. Bua et al.

    Vertical transmission of Trypanosoma cruzi infection: quantification of parasite burden in mothers and their children by parasite DNA amplification

    Trans. R. Soc. Trop. Med. Hyg.

    (2012)
  • J.M. Burgos et al.

    Direct molecular profiling of minicircle signatures and lineages of Trypanosoma cruzi bloodstream populations causing congenital Chagas disease

    Int. J. Parasitol.

    (2007)
  • C.A. Buscaglia et al.

    Tandem amino acid repeats from Trypanosoma cruzi shed antigens increase the half-life of proteins in blood

    Blood

    (1999)
  • C.A. Buscaglia et al.

    The surface coat of the mammal-dwelling infective trypomastigote stage of Trypanosoma cruzi is formed by highly diverse immunogenic mucins

    J. Biol. Chem.

    (2004)
  • S. Buus et al.

    High-resolution mapping of linear antibody epitopes using ultrahigh-density peptide microarrays

    Mol. Cell Proteomics

    (2012)
  • V. Campo et al.

    Differential accumulation of mutations localized in particular domains of the mucin genes expressed in the vertebrate host stage of Trypanosoma cruzi

    Mol. Biochem. Parasitol.

    (2004)
  • V.A. Campo et al.

    Immunocharacterization of the mucin-type proteins from the intracellular stage of Trypanosoma cruzi

    Microbes Infect

    (2006)
  • Y. Carlier et al.

    Congenital Chagas disease as an ecological model of interactions between Trypanosoma cruzi parasites, pregnant women, placenta and fetuses

    Acta Trop.

    (2015)
  • S.J. Carmona et al.

    Towards high-throughput immunomics for infectious diseases: use of next-generation peptide microarrays for rapid discovery and mapping of antigenic determinants

    Mol. Cell Proteomics

    (2015)
  • C. Castillo et al.

    Phospholipase C gamma and ERK1/2 mitogen activated kinase pathways are differentially modulated by Trypanosoma cruzi during tissue invasion in human placenta

    Exp. Parasitol.

    (2013)
  • G.C. Cerqueira et al.

    Sequence diversity and evolution of multigene families in Trypanosoma cruzi

    Mol. Biochem. Parasitol.

    (2008)
  • R.S. Corral et al.

    Presence of IgM antibodies to Trypanosoma cruzi urinary antigen in sera from patients with acute Chagas' disease

    Int. J. Parasitol.

    (1998)
  • M.E. Cortina et al.

    Electrochemical magnetic microbeads-based biosensor for point-of-care serodiagnosis of infectious diseases

    Biosens. Bioelectron.

    (2016)
  • J.R. Coura et al.

    Emerging Chagas disease in Amazonian Brazil

    Trends Parasitol.

    (2002)
  • J.F. da Silveira et al.

    Chagas disease: recombinant Trypanosoma cruzi antigens for serological diagnosis

    Trends Parasitol.

    (2001)
  • A. da Silveira Pinto et al.

    Experimental Trypanosoma cruzi biclonal infection in Triatoma infestans: detection of distinct clonal genotypes using kinetoplast DNA probes

    Int. J. Parasitol.

    (2000)
  • M. de Lana et al.

    Trypanosoma cruzi: compared vectorial transmissibility of three major clonal genotypes by Triatoma infestans

    Exp. Parasitol.

    (1998)
  • R.M. de Lederkremer et al.

    Glycobiology of Trypanosoma cruzi

    Adv. Carbohydr. Chem. Biochem.

    (2009)
  • E.H. de Titto et al.

    Serum neuraminidase activity and hematological alterations in acute human Chagas' disease

    Clin. Immunol. Immunopathol.

    (1988)
  • T. Fernandez-Calero et al.

    Profiling of small RNA cargo of extracellular vesicles shed by Trypanosoma cruzi reveals a specific extracellular signature

    Mol. Biochem. Parasitol.

    (2015)
  • A.C. Frasch

    Functional diversity in the trans-sialidase and mucin families in Trypanosoma cruzi

    Parasitol. Today

    (2000)
  • A.C. Frasch et al.

    Comparison of genes encoding Trypanosoma cruzi antigens

    Parasitol. Today

    (1991)
  • L.M. Galvao et al.

    Lytic antibody titre as a means of assessing cure after treatment of Chagas disease: a 10 years follow-up study

    Trans. R. Soc. Trop. Med. Hyg.

    (1993)
  • M. Abraham et al.

    Anti-ribosomal-P antibodies in lupus nephritis, neuropsychiatric lupus, lupus hepatitis, and Chagas' disease: promising yet limited in clinical utility

    Rheumatol. Int.

    (2015)
  • A. Abras et al.

    Serological diagnosis of chronic Chagas disease: is it time for a change?

    J. Clin. Microbiol.

    (2016)
  • A.A. Ackermann et al.

    A genomic scale map of genetic diversity in Trypanosoma cruzi

    BMC Genom.

    (2012)
  • F. Aguero et al.

    Genomic-scale prioritization of drug targets: the TDR Targets database

    Nat. Rev. Drug Discov.

    (2008)
  • B. Alarcon de Noya et al.

    Large urban outbreak of orally acquired acute Chagas disease at a school in Caracas, Venezuela

    J. Infect. Dis.

    (2010)
  • M.C. Albareda et al.

    Modulation of Trypanosoma cruzi-specific T-cell responses after chemotherapy for chronic Chagas disease

    Mem. Inst. Oswaldo Cruz

    (2015)
  • M.C. Albareda et al.

    Chronic human infection with Trypanosoma cruzi drives CD4+ T cells to immune senescence

    J. Immunol.

    (2009)
  • I.C. Almeida et al.

    A highly sensitive and specific chemiluminescent enzyme-linked immunosorbent assay for diagnosis of active Trypanosoma cruzi infection

    Transfusion

    (1997)
  • M.G. Alvarez et al.

    Treatment success in Trypanosoma cruzi infection is predicted by early changes in serially monitored parasite-specific T and B Cell responses

    PLoS Negl. Trop. Dis.

    (2016)
  • P. Alvarez et al.

    Multiple overlapping epitopes in the repetitive unit of the shed acute-phase antigen from Trypanosoma cruzi enhance its immunogenic properties

    Infect. Immun.

    (2001)
  • A.L. Andrade et al.

    Short report: benznidazole efficacy among Trypanosoma cruzi-infected adolescents after a six-year follow-up

    Am. J. Trop. Med. Hyg.

    (2004)
  • S.G. Andrade

    Influence of Trypanosoma cruzi strain on the pathogenesis of chronic myocardiopathy in mice

    Mem. Inst. Oswaldo Cruz

    (1990)
  • C. Anish et al.

    Immunogenicity and diagnostic potential of synthetic antigenic cell surface glycans of Leishmania

    ACS Chem. Biol.

    (2013)
  • P.R. Antas et al.

    A reliable and specific enzyme-linked immunosorbent assay for the capture of IgM from human Chagasic sera using fixed epimastigotes of Trypanosoma cruzi

    Parasitol. Res.

    (2000)
  • F.G. Araujo

    Analysis of Trypanosoma cruzi antigens bound by specific antibodies and by antibodies to related trypanosomatids

    Infect. Immun.

    (1986)
  • C.M. Ayo et al.

    Genetic susceptibility to Chagas disease: an overview about the infection and about the association between disease and the immune response genes

    Biomed. Res. Int.

    (2013)
  • V. Balouz et al.

    Mapping antigenic motifs in the trypomastigote small surface antigen from Trypanosoma cruzi

    Clin. Vaccine Immunol.

    (2015)
  • Cited by (0)

    View full text