Skip to main content
Advertisement
  • Loading metrics

Ten years of Chagas disease research: Looking back to achievements, looking ahead to challenges

  • Eric Dumonteil ,

    edumonte@tulane.edu

    Affiliation Department of Tropical Medicine, Vector-Borne Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America

  • Claudia Herrera

    Affiliation Department of Tropical Medicine, Vector-Borne Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America

Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, is a neglected tropical disease (NTD) with a high disease burden in the Americas. It is transmitted primarily by hematophagous triatomine bugs, but alternate transmission routes such as congenital, oral, and transfusional transmission are becoming more relevant in many regions. After a short acute phase, infected patients enter an asymptomatic chronic phase, which can become symptomatic in 20% to 40% of the cases, characterized by a chronic chagasic cardiomyopathy and less frequently by a digestive form of the disease. Current control is mostly focused on vector control with indoor spraying of residual pyrethroids and to a lesser extent with housing improvement. Treatment of infected patients remains challenging due to the limited efficacy of the two available drugs during the chronic phase and their side effects but also because of limited access to treatment for patients.

Over the past ten years, PLOS Neglected Tropical Diseases has been a key journal for the diffusion of some of the major achievements to better understand and control Chagas disease. With a total of 390 published studies during that time, Chagas disease research represented about 8% of the published material by the journal. Some of the key issues addressed have been related to the evaluation of disease burden, improvement in serological and molecular diagnostics, drug development, patient care, and vector control (Table 1).

thumbnail
Table 1. Challenges and research priorities for Chagas disease.

https://doi.org/10.1371/journal.pntd.0005422.t001

New estimates of Chagas disease burden and its epidemiological impact provide the basis for health interventions in both endemic and nonendemic countries. Indeed, Chagas disease is responsible for one of the largest disease burden in the Americas where it is endemic, with over 6 million cases. It also has the peculiarity of being one of the few NTDs to cause most of its burden in upper-middle income countries [1], including in the United States where an estimated 300,000 cases are present, and a growing number of autochthonous cases are being identified [2,3]. In nonendemic regions such as in Europe, Chagas disease is a growing concern as well, with an estimated 68,000–120,000 patients [4]. However, the identification of infected patients remains challenging in many countries, and underreporting is still a major issue.

Contrary to most, if not all, other infectious diseases, at least two serological tests are still needed for a reliable serological diagnostic of T. cruzi infection, and additional tests need to be performed in case of discordance among tests. Cases of individuals who are seronegative with conventional serological tests but seropositive with alternative tests or T. cruzi PCR-positive have been reported [5]. Part of the discordances may be attributed to the very large genetic and antigenic diversity of T. cruzi, which has been divided into seven discrete typing units (DTUs) TcI–TcVI and Tcbat [6,7]. Current serological tests are thus based on limited sets of parasite antigens and do not reflect the entire range of diversity of parasite strains and DTUs infecting humans. While efforts have been made to identify novel parasite antigens for serological diagnostic [8], an ideal test is still urgently needed [9,10]. Improved serological tests would allow a better epidemiologic surveillance for the early detection of vectorial, congenital, and oral cases, as well as for the prevention of transfusional cases.

Molecular diagnostic by PCR and quantitative PCR (qPCR) has been refined and standardized, providing key tools for the specific detection, quantification, and genotyping of T. cruzi parasites in a variety of biological and clinical samples [1113]. Such methods are proving critical to complement serological diagnostics, patient follow-up after drug treatment, and the unraveling of parasite transmission cycles. However, the use of PCR as a diagnostic tool is still debated, as the extreme sensitivity that has been reached is prone to false positive results and, most importantly, parasite DNA may be present without infection with live parasites, as it has been observed during congenital infection in mouse models [14,15]. These issues complicate the interpretation of PCR results. On the other hand, molecular genotyping is still insufficiently sensitive, particularly for samples with very low parasite burden [16]. Thus, linking parasite genotypes and DTUs with their biological characteristics, clinical outcomes, and transmission cycles remains elusive [17], and more sensitive genotyping tests are thus critically needed.

In spite of limited options, drug treatment of infected patients has also progressed over the past ten years. Nifurtimox and benznidazole have a confirmed efficacy in the early stages of infection, particularly in children and young adults. Although more studies are needed, the treatment of women of child-bearing age may also prevent congenital transmission to their newborns [18,19]. Importantly, a pediatric formulation of benznidazole has been developed under the leadership of Drugs for Neglected Diseases Initiative (DNDi), making drug administration to children easier (Clinicaltrial.gov registration: NCT02625974). However, drug shortage, lack of registration in many countries, poor access to health care, and limited awareness of the disease in both patients and health providers all reduce access to treatment for patients. Current estimates suggest that only 1% have access to these trypanocidal drugs [20,21]. Eliminating the barriers limiting access to treatment is thus a key priority for the forthcoming years, and this will require strong political commitment.

Nonetheless, the results of the long awaited Benznidazole Evaluation for Interrupting Trypanosomiasis (BENEFIT) trial, aimed at evaluating benznidazole treatment in patients with chronic chagasic cardiomyopathy, have stressed the limitations of current treatment [22]. Indeed, while treated patients presented significant decreases in blood parasite levels, they showed no improvement in cardiac clinical outcomes [23]. In addition, the development of new drugs is progressing at a disappointing rate, and there are very few candidates in the development pipeline [21]. Posaconazole, once viewed as a very promising new option, failed to provide sustained reduction in circulating parasites in clinical trials, and other triazoles (i.e., ravuconazole) are not superior to benznidazole [24]. As an alternative, vaccine development against T. cruzi has benefited from a renewed interest [25] due to a better understanding of the delicate host–parasite immune balance, and vaccine development may lead to a valuable additional tool for Chagas disease control.

These recent clinical trials have also pointed out the need for a better understanding of T. cruzi pathogenesis and disease progression. Indeed, the assessment of blood parasite burden by qPCR or the reduction/disappearance of antibodies against the parasite are very imperfect end points to evaluate treatment effectiveness. New biomarkers of disease progression and treatment efficacy are urgently needed for a better follow-up of treated patients. Several candidate molecules have been described [26] but still require extensive clinical validation, which can only be achieved through the development of easy and affordable assays. The dynamics of parasite tissue distribution and the multiclonality of infections are also just beginning to be addressed and may lead to a better understanding of the delicate balance in the host-parasite relationship [27].

As mentioned above, vector control remains the main preventive intervention against Chagas disease in endemic countries, and it has been very effective at eliminating vectorial transmission by triatomine species that have adapted well to human housing such as Triatoma infestans [28]. Thus, several countries and regions have been declared free of vectorial transmission in the past decade [29]. However, many additional species of triatomines that are more sylvatic but with various degrees of intrusiveness to human habitat are now emerging as important vectors responsible for a significant transmission of T. cruzi to humans [28]. Their control with conventional indoor residual insecticide spraying is poorly effective and thus requires the development of novel integrative interventions based on triatomine ecology. Entomological surveillance and control thus need to be further strengthened and sustained to achieve better vector control.

While we have witnessed striking progress over the past ten years in our understanding and control of Chagas disease, the major challenges highlighted above remain to be addressed to reach the goals of the London declaration on NTD, which aims at eliminating this disease (among others) as a public health concern. There is no doubt that PLOS Neglected Tropical Diseases will continue to support this endeavor by providing a forum for the discussion of these challenges and our progress in addressing them.

References

  1. 1. Stolk WA, Kulik MC, le Rutte EA, Jacobson J, Richardus JH, et al. (2016) Between-Country Inequalities in the Neglected Tropical Disease Burden in 1990 and 2010, with Projections for 2020. PLoS Negl Trop Dis 10: e0004560. pmid:27171193
  2. 2. Hotez PJ, Dumonteil E, Betancourt Cravioto M, Bottazzi ME, Tapia-Conyer R, et al. (2013) An unfolding tragedy of chagas disease in north America. PLoS Negl Trop Dis 7: e2300. pmid:24205411
  3. 3. Garcia MN, Aguilar D, Gorchakov R, Rossmann SN, Montgomery SP, et al. (2015) Evidence of autochthonous Chagas disease in southeastern Texas. Am J Trop Med Hyg 92: 325–330. pmid:25371187
  4. 4. Requena-Mendez A, Aldasoro E, de Lazzari E, Sicuri E, Brown M, et al. (2015) Prevalence of Chagas disease in Latin-American migrants living in Europe: a systematic review and meta-analysis. PLoS Negl Trop Dis 9: e0003540. pmid:25680190
  5. 5. Salomone OA, Basquiera AL, Sembaj A, Aguerri AM, Reyes ME, et al. (2003) Trypanosoma cruzi in persons without serologic evidence of disease, Argentina. Emerg Infect Dis 9: 1558–1562. pmid:14720396
  6. 6. Zingales B, Miles MA, Campbell DA, Tibayrenc M, Macedo AM, et al. (2012) The revised Trypanosoma cruzi subspecific nomenclature: rationale, epidemiological relevance and research applications. Infect Genet Evol 12: 240–253. pmid:22226704
  7. 7. Breniere SF, Waleckx E, Barnabe C (2016) Over Six Thousand Trypanosoma cruzi Strains Classified into Discrete Typing Units (DTUs): Attempt at an Inventory. PLoS Negl Trop Dis 10: e0004792. pmid:27571035
  8. 8. Cooley G, Etheridge RD, Boehlke C, Bundy B, Weatherly DB, et al. (2008) High throughput selection of effective serodiagnostics for Trypanosoma cruzi infection. PLoS Negl Trop Dis 2: e316. pmid:18841200
  9. 9. Afonso AM, Ebell MH, Tarleton RL (2012) A systematic review of high quality diagnostic tests for Chagas disease. PLoS Negl Trop Dis 6: e1881. pmid:23145201
  10. 10. Porras AI, Yadon ZE, Altcheh J, Britto C, Chaves GC, et al. (2015) Target Product Profile (TPP) for Chagas Disease Point-of-Care Diagnosis and Assessment of Response to Treatment. PLoS Negl Trop Dis 9: e0003697. pmid:26042730
  11. 11. Duffy T, Bisio M, Altcheh J, Burgos JM, Diez M, et al. (2009) Accurate real-time PCR strategy for monitoring bloodstream parasitic loads in chagas disease patients. PLoS Negl Trop Dis 3: e419. pmid:19381287
  12. 12. Wei B, Chen L, Kibukawa M, Kang J, Waskin H, et al. (2016) Development of a PCR Assay to Detect Low Level Trypanosoma cruzi in Blood Specimens Collected with PAXgene Blood DNA Tubes for Clinical Trials Treating Chagas Disease. PLoS Negl Trop Dis 10: e0005146. pmid:27906977
  13. 13. Ramirez JC, Cura CI, da Cruz Moreira O, Lages-Silva E, Juiz N, et al. (2015) Analytical Validation of Quantitative Real-Time PCR Methods for Quantification of Trypanosoma cruzi DNA in Blood Samples from Chagas Disease Patients. J Mol Diagn 17: 605–615. pmid:26320872
  14. 14. Cencig S, Coltel N, Truyens C, Carlier Y (2013) Fertility, gestation outcome and parasite congenital transmissibility in mice infected with TcI, TcII and TcVI genotypes of Trypanosoma cruzi. PLoS Negl Trop Dis 7: e2271. pmid:23785533
  15. 15. Carlier Y, Torrico F, Sosa-Estani S, Russomando G, Luquetti A, et al. (2011) Congenital Chagas disease: recommendations for diagnosis, treatment and control of newborns, siblings and pregnant women. PLoS Negl Trop Dis 5: e1250. pmid:22039554
  16. 16. Garcia MN, Burroughs H, Gorchakov R, Gunter SM, Dumonteil E, et al. (2017) Molecular identification and genotyping of Trypanosoma cruzi DNA in autochthonous Chagas disease patients from Texas, USA. Infect Genet Evol 49: 151–156. pmid:28095298
  17. 17. Messenger LA, Miles MA, Bern C (2015) Between a bug and a hard place: Trypanosoma cruzi genetic diversity and the clinical outcomes of Chagas disease. Expert Rev Anti Infect Ther 13: 995–1029. pmid:26162928
  18. 18. Moscatelli G, Moroni S, Garcia-Bournissen F, Ballering G, Bisio M, et al. (2015) Prevention of congenital Chagas through treatment of girls and women of childbearing age. Mem Inst Oswaldo Cruz 110: 507–509. pmid:25993401
  19. 19. Fabbro DL, Danesi E, Olivera V, Codebo MO, Denner S, et al. (2014) Trypanocide treatment of women infected with Trypanosoma cruzi and its effect on preventing congenital Chagas. PLoS Negl Trop Dis 8: e3312. pmid:25411847
  20. 20. Manne JM, Snively CS, Ramsey JM, Salgado MO, Barnighausen T, et al. (2013) Barriers to treatment access for Chagas disease in Mexico. PLoS Negl Trop Dis 7: e2488. pmid:24147169
  21. 21. Ribeiro I, Sevcsik AM, Alves F, Diap G, Don R, et al. (2009) New, improved treatments for chagas disease: from the R&D pipeline to the patients. PLoS Negl Trop Dis 3: e484. pmid:19582163
  22. 22. Pecoul B, Batista C, Stobbaerts E, Ribeiro I, Vilasanjuan R, et al. (2016) The BENEFIT Trial: Where Do We Go from Here? PLoS Negl Trop Dis 10: e0004343. pmid:26913759
  23. 23. Morillo CA, Marin-Neto JA, Avezum A, Sosa-Estani S, Rassi A Jr., et al. (2015) Randomized Trial of Benznidazole for Chronic Chagas' Cardiomyopathy. N Engl J Med 373: 1295–1306. pmid:26323937
  24. 24. Urbina JA (2015) Recent clinical trials for the etiological treatment of chronic chagas disease: advances, challenges and perspectives. J Eukaryot Microbiol 62: 149–156. pmid:25284065
  25. 25. Dumonteil E, Bottazzi ME, Zhan B, Heffernan MJ, Jones K, et al. (2012) Accelerating the development of a therapeutic vaccine for human Chagas disease: rationale and prospects. Expert Rev Vaccines 11: 1043–1055. pmid:23151163
  26. 26. Pinazo MJ, Thomas MC, Bua J, Perrone A, Schijman AG, et al. (2014) Biological markers for evaluating therapeutic efficacy in Chagas disease, a systematic review. Expert Rev Anti Infect Ther 12: 479–496. pmid:24621252
  27. 27. Lewis MD, Kelly JM (2016) Putting Infection Dynamics at the Heart of Chagas Disease. Trends Parasitol 32: 899–911. pmid:27612651
  28. 28. Waleckx E, Gourbière S, Dumonteil E (2015) Intrusive triatomines and the challenge of adapting vector control practices. Mem Inst Oswaldo Cruz 110: 324–338. pmid:25993504
  29. 29. Abad-Franch F, Diotaiuti L, Gurgel-Goncalves R, Gurtler RE (2013) Certifying the interruption of Chagas disease transmission by native vectors: cui bono? Mem Inst Oswaldo Cruz 108: 251–254. pmid:23579810