1
|
Calderón-Quintal JA, Escalante-Talavera MJ, Teh-Poot CF, Carrera-Campellone MN, Martinez-Vega PP, Dzul-Huchim VM, Pech-Pisté LM, Waleckx EB, Villanueva-Lizama LE, Ortega-Lopez J, Dumonteil E, Cruz-Chan JV. Natural infection of Trypanosoma Cruzi in client-owned-dogs from rural Yucatan, Mexico. Sci Rep 2025; 15:10263. [PMID: 40133329 PMCID: PMC11937323 DOI: 10.1038/s41598-025-92176-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Dogs play a crucial role in the transmission cycle of Trypanosoma cruzi and their presence in domicile increases the risk of infection in humans. In Yucatán, Mexico previous studies have reported T. cruzi infection in dogs from both rural and urban areas, which we expanded here, to better understand infection dynamics. A total of 186-dogs were screened for T. cruzi infection by PCR and serology. Parasite burden, genotype, immune response, cardiac alterations, and roaming behavior of the dogs were analyzed. The T. cruzi prevalence was 26.8% (50/186). Genotyping of T. cruzi revealed the predominance of TcI parasites, although most dogs (15/25, 60%) harbored mixed infections with additional DTUs including TcII, TcIV, TcV and TcVI. Antibodies against T. cruzi proteins were detected in > 90% of infected dogs, confirming their immunogenicity in natural infections. Mild ECG abnormalities were present in 40% of infected dogs. A logistic model suggested that the interplay between the host responses to multiple parasite strains could mediate differences in disease severity (P = 0.0002, R2 = 0.65). Finally, parasite diversity and dog roaming behavior support a role of dogs as an important link in T. cruzi transmission cycles among habitats. Together, these data provide a strong rationale to target dogs in integrated Chagas disease control interventions.
Collapse
Affiliation(s)
- Jorge Andrés Calderón-Quintal
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
| | - Maryrose José Escalante-Talavera
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
| | - Christian Florian Teh-Poot
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
| | - María Noe Carrera-Campellone
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
| | - Pedro Pablo Martinez-Vega
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
| | - Victor Manuel Dzul-Huchim
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
| | - Landy Magaly Pech-Pisté
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
| | - Etienne B Waleckx
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México
- Institut de Recherche pour le Développment, UMR INTERTRYP IRD, CIRAD, Université de Montpellier, Montpellier, France
| | | | - Jaime Ortega-Lopez
- Departamento de Biotecnología y Bioingeniería, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, México
| | - Eric Dumonteil
- Department of Tropical Medicine and Infectious Disease, and Vector-Borne and Infectious Disease Research Center, Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, United States.
| | - Julio Vladimir Cruz-Chan
- Laboratorio de Parasitología, Centro de Investigaciones Regionales "Dr Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, México.
| |
Collapse
|
2
|
Simões-Neto EA, Santos DWDCL, Bomfim MRQ, Costa JML, Simões AF, Vasconcelos LD, Sodré DC, Costa ACM, Dumont SVR, de Oliveira de Melo B, de Azevedo CDMPES. Oral Chagas disease outbreak by bacaba juice ingestion: A century after Carlos Chagas' discovery, the disease is still hard to manage. PLoS Negl Trop Dis 2024; 18:e0012225. [PMID: 39292716 PMCID: PMC11441692 DOI: 10.1371/journal.pntd.0012225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 09/30/2024] [Accepted: 07/24/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Orally transmitted acute Chagas disease (ACD) primarily affects low-visibility and low-income individuals in tropical and subtropical zones. Managing ACD remains challenging even after more than 100 years of its discovery. Its spread to non-endemic areas has made it a global health issue. The aim of this work is to demonstrate the difficulties encountered in handling a real-life situation. METHODOLOGY AND FINDINGS This report examines an outbreak of 39 cases of ACD due to oral transmission by bacaba juice ingestion that occurred in Pedro do Rosário, Maranhão, Brazil. A clinical and epidemiological investigation, including an entomological search, was conducted. Diagnosis criteria included positive peripheral blood smear (PBS), seroconversion of IgG, and a two-fold increase in IgG titer (laboratory criteria); and clinical findings, epidemiological exposure, and at least one positive IgG test (clinical-epidemiological criteria). In-house conventional polymerase chain reaction (PCR) was performed on 33 samples. All patients were treated with benznidazole. After 4.5 years, IgG levels were reassessed in 26 individuals. The mean age was 33.6 years, with no gender difference. The mean incubation period was 13.8 days, and the mean between symptom onset and treatment was 16.6 days. The most common symptoms were fever and lymphadenopathy (90%). Diagnostic success rates were 66.6% (laboratory criteria), 23% (clinical-epidemiological criteria), and 10.2% (high clinical suspicion despite negative tests). Test positivity rates were 69.7% (PBS), 91.4% (serology), and 100% (PCR). There were no deaths. Serological cure was achieved in 34.6% of cases, and IgG titers decreased in 15.3%. CONCLUSIONS AND SIGNIFICANCE We encountered several barriers in managing ACD, including population vulnerability, reliance on outdated diagnostic techniques, lack of standardized molecular biology methods, and limited therapeutic options. This report underscores the importance of rapid surveillance and early treatment to prevent fatalities. We recommend the standardization of conventional PCR in diagnostic routines.
Collapse
Affiliation(s)
| | - Daniel Wagner de Castro Lima Santos
- Presidente Dutra University Hospital (HU-UPD), Brazilian Company of Hospital Services (EBSERH), Federal University of Maranhão (UFMA), São Luís, Brazil
- IDOR, Instituto D'Or de Pesquisa e Ensino
| | | | | | - Amanda Ferreira Simões
- Brasília University Hospital (HUB), Brazilian Company of Hospital Services (EBSERH), Brasília University (UnB), Brasília, Brazil
| | | | | | | | | | | | | |
Collapse
|
3
|
Ramos LG, de Souza KR, Júnior PAS, Câmara CC, Castelo-Branco FS, Boechat N, Carvalho SA. Tackling the challenges of human Chagas disease: A comprehensive review of treatment strategies in the chronic phase and emerging therapeutic approaches. Acta Trop 2024; 256:107264. [PMID: 38806090 DOI: 10.1016/j.actatropica.2024.107264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024]
Abstract
Chagas disease (CD), caused by the flagellated protozoan Trypanosoma cruzi (T. cruzi), affects approximately 7 million people worldwide and is endemic in Latin America, especially among socioeconomically disadvantaged populations. Since the 1960s, only two drugs have been commercially available for treating this illness: nifurtimox (NFX) and benznidazole (BZN). Although these drugs are effective in the acute phase (AP) of the disease, in which parasitemia is usually high, their cure rates in the chronic phase (CP) are low and often associated with several side effects. The CP is characterized by a subpatent parasitaemia and absence of clinical symptoms in the great majority of infected individuals. However, at least 30 % of the individuals will develop potentially lethal symptomatic forms, including cardiac and digestive manifestations. For such reason, in the CP the treatment is usually symptomatic and typically focuses on managing complications such as arrhythmias, heart failure, or digestive problems. Therefore, the need for new drugs or therapeutic approaches using BZN or NFX is extremely urgent. This review presents the main clinical trials, especially in the CP, which involve BZN and NFX in different treatment regimens. Additionally, other therapies using combinations of these drugs with other substances such as allopurinol, itraconazole, ravuconazole, ketoconazole, posaconazole and amiodarone are also reported. The importance of early diagnosis, especially in pediatric patients, is also discussed, emphasizing the need to identify the disease in its early stages to improve the chances of successful treatment.
Collapse
Affiliation(s)
- Laís Gomes Ramos
- Laboratorio de Sintese de Farmacos -LASFAR, Instituto de Tecnologia em Farmacos - Farmanguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, RJ 21041-250, Brazil; Laboratório de Físico-Química de Materiais, Seção de Engenharia Química, Instituto Militar de Engenharia, Praça General Tibúrcio 80, Rio de Janeiro, RJ 22290-270, Brazil
| | - Kátia Regina de Souza
- Laboratório de Físico-Química de Materiais, Seção de Engenharia Química, Instituto Militar de Engenharia, Praça General Tibúrcio 80, Rio de Janeiro, RJ 22290-270, Brazil
| | - Policarpo Ademar Sales Júnior
- Laboratório de Imunopatologia e Biologia Molecular, Departamento de Imunologia, Instituto Ageu Magalhães, Fundação Oswaldo Cruz, Recife, PE 50670-420, Brazil
| | - Camila Capelini Câmara
- Laboratorio de Sintese de Farmacos -LASFAR, Instituto de Tecnologia em Farmacos - Farmanguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, RJ 21041-250, Brazil; Laboratório de Físico-Química de Materiais, Seção de Engenharia Química, Instituto Militar de Engenharia, Praça General Tibúrcio 80, Rio de Janeiro, RJ 22290-270, Brazil
| | - Frederico S Castelo-Branco
- Laboratorio de Sintese de Farmacos -LASFAR, Instituto de Tecnologia em Farmacos - Farmanguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, RJ 21041-250, Brazil
| | - Nubia Boechat
- Laboratorio de Sintese de Farmacos -LASFAR, Instituto de Tecnologia em Farmacos - Farmanguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, RJ 21041-250, Brazil
| | - Samir Aquino Carvalho
- Laboratorio de Sintese de Farmacos -LASFAR, Instituto de Tecnologia em Farmacos - Farmanguinhos, Fundação Oswaldo Cruz, Rio de Janeiro, RJ 21041-250, Brazil.
| |
Collapse
|
4
|
Farani PSG, Jones KM, Poveda C. Treatments and the Perspectives of Developing a Vaccine for Chagas Disease. Vaccines (Basel) 2024; 12:870. [PMID: 39203996 PMCID: PMC11359273 DOI: 10.3390/vaccines12080870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Chagas disease (CD) treatment and vaccine development are critical due to the significant health burden caused by the disease, especially in Latin America. Current treatments include benznidazole and nifurtimox, which are most effective in the acute phase of the disease but less so in the chronic phase, often with significant side effects. Here, using the available literature, we summarize the progress in vaccine development and new treatments that promise to reduce CD incidence and improve the quality of life for those at risk, particularly in endemic regions. New treatment options, such as posaconazole and fexinidazole, are being explored to improve efficacy and reduce adverse effects. Vaccine development for CD remains a high priority. The complex life stages and genetic diversity of Trypanosoma cruzi present challenges, but several promising vaccine candidates are under investigation. These efforts focus on stimulating a protective immune response through various innovative approaches.
Collapse
Affiliation(s)
- Priscila Silva Grijó Farani
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Kathryn Marie Jones
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristina Poveda
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Torchelsen FKVDS, Fernandes TCP, de Sousa SMR, Sales-Junior PA, Branquinho RT, Murta SMF, Teixeira RR, Mosqueira VCF, de Lana M. Screening of synthetic 1,2,3-triazolic compounds inspired by SRPIN340 as anti-Trypanosoma cruzi agents. Rev Soc Bras Med Trop 2024; 57:e00411. [PMID: 39082521 PMCID: PMC11290850 DOI: 10.1590/0037-8682-0585-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 05/09/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND The current treatments for Chagas disease (CD) include benznidazole and nifurtimox, which have limited efficacy and cause numerous side effects. Triazoles are candidates for new CD treatments due to their ability to eliminate T. cruzi parasites by inhibiting ergosterol synthesis, thereby damaging the cell membranes of the parasite. METHODS Eleven synthetic analogs of the kinase inhibitor SRPIN340 containing a triazole core (compounds 6A-6K) were screened in vitro against the Tulahuen strain transfected with β-galactosidase, and their IC50, CC50, and selectivity indexes (SI) were calculated. Compounds with an SI > 50 were further evaluated in mice infected with the T. cruzi Y strain by rapid testing. RESULTS Eight compounds were active in vitro with IC50 values ranging from 0.5-10.5 µg/mL. The most active compounds, 6E and 6H, had SI values of 125.2 and 69.6, respectively. These compounds also showed in vivo activity, leading to a reduction in parasitemia at doses of 10, 50, and 250 mg/kg/day. At doses of 50 and 250 mg/kg/day, parasitemia was significantly reduced compared to infected untreated animals, with no significant differences between the effects of 6E and 6H. CONCLUSIONS This study identified two new promising compounds for CD chemotherapy and confirmed their activity against T. cruzi.
Collapse
Affiliation(s)
| | - Tamiles Caroline Pedrosa Fernandes
- Universidade Federal de Ouro Preto, Programa de Pós-Graduação em
Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Ouro Preto, MG,
Brasil
| | | | | | - Renata Tupinambá Branquinho
- Universidade Federal de Ouro Preto, Programa de Pós-Graduação em
Ciências Farmacêuticas, Ouro Preto, MG, Brasil
| | | | | | | | - Marta de Lana
- Universidade Federal de Ouro Preto, Programa de Pós-Graduação em
Ciências Farmacêuticas, Ouro Preto, MG, Brasil
- Universidade Federal de Ouro Preto, Programa de Pós-Graduação em
Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Ouro Preto, MG,
Brasil
| |
Collapse
|
6
|
Torchelsen FKVDS, Mazzeti AL, Mosqueira VCF. Drugs in preclinical and early clinical development for the treatment of Chagas´s disease: the current status. Expert Opin Investig Drugs 2024; 33:575-590. [PMID: 38686546 DOI: 10.1080/13543784.2024.2349289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Chagas disease is spreading faster than expected in different countries, and little progress has been reported in the discovery of new drugs to combat Trypanosoma cruzi infection in humans. Recent clinical trials have ended with small hope. The pathophysiology of this neglected disease and the genetic diversity of parasites are exceptionally complex. The only two drugs available to treat patients are far from being safe, and their efficacy in the chronic phase is still unsatisfactory. AREAS COVERED This review offers a comprehensive examination and critical review of data reported in the last 10 years, and it is focused on findings of clinical trials and data acquired in vivo in preclinical studies. EXPERT OPINION The in vivo investigations classically in mice and dog models are also challenging and time-consuming to attest cure for infection. Poorly standardized protocols, availability of diagnosis methods and disease progression markers, the use of different T. cruzi strains with variable benznidazole sensitivities, and animals in different acute and chronic phases of infection contribute to it. More synchronized efforts between research groups in this field are required to put in evidence new promising substances, drug combinations, repurposing strategies, and new pharmaceutical formulations to impact the therapy.
Collapse
Affiliation(s)
- Fernanda Karoline Vieira da Silva Torchelsen
- School of Pharmacy, Federal University of Ouro Preto, Ouro Preto, Brazil
- Post-Graduation Program in Pharmaceutical Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ana Lia Mazzeti
- Department of Biomedical Sciences and Health, Academic Unit of Passos, University of Minas Gerais State, Passos, Brazil
| | | |
Collapse
|
7
|
de Sousa AS, Vermeij D, Ramos AN, Luquetti AO. Chagas disease. Lancet 2024; 403:203-218. [PMID: 38071985 DOI: 10.1016/s0140-6736(23)01787-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 01/15/2024]
Abstract
Chagas disease persists as a global public health problem due to the high morbidity and mortality burden. Despite the possibility of a cure and advances in transmission control, epidemiological transformations, such as urbanisation and globalisation, and the emerging importance of oral and vertical transmission mean that Chagas disease should be considered an emerging disease, with new cases occurring worldwide. Important barriers to diagnosis, treatment, and care remain, resulting in repressed numbers of reported cases, which in turn leads to inadequate public policies. The validation of new diagnostic tools and treatment options is needed, as existing tools pose serious limitations to access to health care. Integrated models of surveillance, with community and intersectional participation, embedded in the concept of One Health, are essential for control. In addition, mitigation strategies for the main social determinants of health, including difficulties imposed by migration, are important to improve access to comprehensive health care in a globalised scenario.
Collapse
Affiliation(s)
- Andréa Silvestre de Sousa
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Department of Internal Medicine, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Debbie Vermeij
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Alberto Novaes Ramos
- Department of Community Health, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Alejandro O Luquetti
- Center of Studies for Chagas Disease, Hospital das Clínicas, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
8
|
Moscatelli G, Moroni S, Ramírez JC, Warszatska B, Fernanda L, González N, Rabinovich A, Altcheh J. Efficacy of short-course treatment for prevention of congenital transmission of Chagas disease: A retrospective cohort study. PLoS Negl Trop Dis 2024; 18:e0011895. [PMID: 38252673 PMCID: PMC10833510 DOI: 10.1371/journal.pntd.0011895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/01/2024] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND In regions with controlled vector transmission of T. cruzi, congenital transmission is the most frequent route of infection. Treatment with benznidazole (BZ) or nifurtimox (NF) for 60 days in girls and women of childbearing age showed to be effective in preventing mother to child transmission of this disease. Reports on short-course treatment (≤30 days) are scarce. METHODS Retrospective cohort study. Offspring of women with Chagas disease who received short-course treatment (≤30 days) with BZ or NF, attended between 2003 and 2022, were evaluated. Parasitemia (microhaematocrit and/or PCR) was performed at <8 months of age, and serology (ELISA and IHA) at ≥8 months to rule out congenital infection. RESULTS A total of 27 women receiving ≤30 days of treatment and their children were included in this study. NF was prescribed in 17/27 (63%) women, and BZ in 10/27 (37%). The mean duration of treatment was 29.2 days. None of the women experienced serious adverse events during treatment, and no laboratory abnormalities were observed. Forty infants born to these 27 treated women were included. All newborns were full term, with appropriate weight for their gestational age. No perinatal infectious diseases or complications were observed. DISCUSSION Several studies have shown that treatment of infected girls and women of childbearing age for 60 days is an effective practice to prevent transplacental transmission of T. cruzi. Our study demonstrated that short-duration treatment (≤30 days) is effective and beneficial in preventing transplacental transmission of Chagas disease.
Collapse
Affiliation(s)
- Guillermo Moscatelli
- Servicio de Parasitología y Chagas, Hospital de Niños “Ricardo Gutiérrez”, Buenos Aires, Argentina
- Instituto Multidisciplinario de Investigaciones en Patologías Pediatricas (IMIPP)- CONICET-GCBA, Buenos Aires, Argentina
| | - Samanta Moroni
- Servicio de Parasitología y Chagas, Hospital de Niños “Ricardo Gutiérrez”, Buenos Aires, Argentina
- Instituto Multidisciplinario de Investigaciones en Patologías Pediatricas (IMIPP)- CONICET-GCBA, Buenos Aires, Argentina
| | - Juan Carlos Ramírez
- Instituto Multidisciplinario de Investigaciones en Patologías Pediatricas (IMIPP)- CONICET-GCBA, Buenos Aires, Argentina
| | - Belén Warszatska
- Servicio de Parasitología y Chagas, Hospital de Niños “Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Lascano Fernanda
- Servicio de Parasitología y Chagas, Hospital de Niños “Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Nicolás González
- Servicio de Parasitología y Chagas, Hospital de Niños “Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Andrés Rabinovich
- Instituto Multidisciplinario de Investigaciones en Patologías Pediatricas (IMIPP)- CONICET-GCBA, Buenos Aires, Argentina
| | - Jaime Altcheh
- Servicio de Parasitología y Chagas, Hospital de Niños “Ricardo Gutiérrez”, Buenos Aires, Argentina
- Instituto Multidisciplinario de Investigaciones en Patologías Pediatricas (IMIPP)- CONICET-GCBA, Buenos Aires, Argentina
| |
Collapse
|
9
|
Gonzaga BMDS, Ferreira RR, Coelho LL, Carvalho ACC, Garzoni LR, Araujo-Jorge TC. Clinical trials for Chagas disease: etiological and pathophysiological treatment. Front Microbiol 2023; 14:1295017. [PMID: 38188583 PMCID: PMC10768561 DOI: 10.3389/fmicb.2023.1295017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/27/2023] [Indexed: 01/09/2024] Open
Abstract
Chagas disease (CD) is caused by the flagellate protozoan Trypanosoma cruzi. It is endemic in Latin America. Nowadays around 6 million people are affected worldwide, and 75 million are still at risk. CD has two evolutive phases, acute and chronic. The acute phase is mostly asymptomatic, or presenting unspecific symptoms which makes it hard to diagnose. At the chronic phase, patients can stay in the indeterminate form or develop cardiac and/or digestive manifestations. The two trypanocide drugs available for the treatment of CD are benznidazole (BZ) and nifurtimox (NFX), introduced in the clinic more than five decades ago. WHO recommends treatment for patients at the acute phase, at risk of congenital infection, for immunosuppressed patients and children with chronic infection. A high cure rate is seen at the CD acute phase but better treatment schemes still need to be investigated for the chronic phase. There are some limitations within the use of the trypanocide drugs, with side effects occurring in about 40% of the patients, that can lead patients to interrupt treatment. In addition, patients with advanced heart problems should not be treated with BZ. This is a neglected disease, discovered 114 years ago that still has no drug effective for their chronic phase. Multiple social economic and cultural barriers influence CD research. The high cost of the development of new drugs, in addition to the low economical return, results in the lack of investment. More economic support is required from governments and pharmaceutical companies on the development of more research for CD treatment. Two approaches stand out: repositioning and combination of drugs, witch drastically decrease the cost of this process, when compared to the development of a new drug. Here we discuss the progress of the clinical trials for the etiological and pathophysiological treatment for CD. In summary, more studies are needed to propose a new drug for CD. Therefore, BZ is still the best option for CD. The trials in course should clarify more about new treatment regimens, but it is already possible to indicate that dosage and time of treatment need to be adjusted.
Collapse
Affiliation(s)
| | | | | | | | | | - Tania C. Araujo-Jorge
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos - Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
10
|
Vázquez C, Encalada R, Belmont-Díaz J, Rivera M, Alvarez S, Nogueda-Torres B, Saavedra E. Metabolic control analysis of the transsulfuration pathway and the compensatory role of the cysteine transport in Trypanosoma cruzi. Biosystems 2023; 234:105066. [PMID: 37898397 DOI: 10.1016/j.biosystems.2023.105066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023]
Abstract
Trypanosoma cruzi is the causal agent of American Trypanosomiasis or Chagas Disease in humans. The current drugs for its treatment benznidazole and nifurtimox have inconveniences of toxicity and efficacy; therefore, the search for new therapies continues. Validation through genetic strategies of new drug targets against the parasite metabolism have identified numerous essential genes. Target validation can be further narrowed by applying Metabolic Control Analysis (MCA) to determine the flux control coefficients of the pathway enzymes. That coefficient is a quantitative value that represents the degree in which an enzyme/transporter determines the flux of a metabolic pathway; those with the highest coefficients can be promising drug targets. Previous studies have demonstrated that cysteine (Cys) is a key precursor for the synthesis of trypanothione, the main antioxidant metabolite in the parasite. In this research, MCA was applied in an ex vivo system to the enzymes of the reverse transsulfuration pathway (RTP) for Cys synthesis composed by cystathionine beta synthase (CBS) and cystathionine gamma lyase (CGL). The results indicated that CGL has 90% of the control of the pathway flux. Inhibition of CGL with propargylglycine (PAG) decreased the levels of Cys and trypanothione and depleted those of glutathione in epimastigotes (proliferative stage in the insect vector); these metabolite changes were prevented by supplementing with Cys, suggesting a compensatory role of the Cys transport (CysT). Indeed, Cys supplementation (but not PAG treatment) increased the activity of the CysT in epimastigotes whereas in trypomastigotes (infective stage in mammals) CysT was increased when they were incubated with PAG. Our results suggested that CGL could be a potential drug target given its high control on the RTP flux and its effects on the parasite antioxidant defense. However, the redundant Cys supply pathways in the parasite may require inhibition of the CysT as well. Our findings also suggest differential responses of the Cys supply pathways in different parasite stages.
Collapse
Affiliation(s)
- Citlali Vázquez
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, 14080, Mexico; Posgrado en Ciencias Químico Biológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, 11350, Mexico
| | - Rusely Encalada
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, 14080, Mexico
| | - Javier Belmont-Díaz
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, 14080, Mexico
| | - Moisés Rivera
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, 14080, Mexico
| | - Samantha Alvarez
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, 14080, Mexico
| | - Benjamín Nogueda-Torres
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, 11350, Mexico
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, 14080, Mexico.
| |
Collapse
|
11
|
Schijman AG. Unveiling challenges in real-time PCR strategies for detecting treatment failure: observations from clinical trials on chronic Chagas disease. FRONTIERS IN PARASITOLOGY 2023; 2:1260224. [PMID: 39816840 PMCID: PMC11732123 DOI: 10.3389/fpara.2023.1260224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/04/2023] [Indexed: 01/18/2025]
Abstract
Chagas disease (CD) caused by Trypanosoma cruzi remains a Neglected Tropical Disease with limited access to diagnosis and treatment, particularly for chronically infected patients. Clinical trials are underway to improve treatment using new drugs or different regimens, and Real-Time PCR is used to assess the parasitological response as a surrogate biomarker. However, PCR-based strategies have limitations due to the complex nature of T. cruzi infection. The parasite exhibits asynchronous replication, different strains and clones, and diverse tissue tropism, making it challenging to determine optimal timeline points for monitoring treatment response. This mini-review explores factors that affect PCR-based monitoring and summarizes the endpoints used in clinical trials for detecting treatment failure. Serial sampling and cumulative PCR results may improve sensitivity in detecting parasitemia and treatment failure in these trials.
Collapse
Affiliation(s)
- Alejandro G. Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular Dr Héctor N. Torres (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
12
|
Dumonteil E, Desale H, Tu W, Hernandez-Cuevas N, Shroyer M, Goff K, Marx PA, Herrera C. Intra-host Trypanosoma cruzi strain dynamics shape disease progression: the missing link in Chagas disease pathogenesis. Microbiol Spectr 2023; 11:e0423622. [PMID: 37668388 PMCID: PMC10581044 DOI: 10.1128/spectrum.04236-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 07/10/2023] [Indexed: 09/06/2023] Open
Abstract
Chronic Chagasic cardiomyopathy develops years after infection in 20-40% of patients, but disease progression is poorly understood. Here, we assessed Trypanosoma cruzi parasite dynamics and pathogenesis over a 2.5-year period in naturally infected rhesus macaques. Individuals with better control of parasitemia were infected with a greater diversity of parasite strains compared to those with increasing parasitemia over time. Also, the in vivo parasite multiplication rate decreased with increasing parasite diversity, suggesting competition among strains or a stronger immune response in multiple infections. Significant differences in electrocardiographic (ECG) profiles were observed in Chagasic macaques compared to uninfected controls, suggesting early conduction defects, and changes in ECG patterns over time were observed only in macaques with increasing parasitemia and lower parasite diversity. Disease progression was also associated with plasma fibronectin degradation, which may serve as a biomarker. These data provide a novel framework for the understanding of Chagas disease pathogenesis, with parasite diversity shaping disease progression.IMPORTANCEChagas disease progression remains poorly understood, and patients at increased risk of developing severe cardiac disease cannot be distinguished from those who may remain asymptomatic. Monitoring of Trypanosoma cruzi strain dynamics and pathogenesis over 2-3 years in naturally infected macaques shows that increasing parasite diversity in hosts is detrimental to parasite multiplication and Chagasic cardiomyopathy disease progression. This provides a novel framework for the understanding of Chagas disease pathogenesis.
Collapse
Affiliation(s)
- Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| | - Hans Desale
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| | - Weihong Tu
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| | - Nora Hernandez-Cuevas
- Laboratorio de Parasitologia, Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autonoma de Yucatan, Merida, Yucatan, Mexico
| | - Monica Shroyer
- Division of Veterinary Medicine, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Kelly Goff
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Preston A. Marx
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Claudia Herrera
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
13
|
Marin-Neto JA, Rassi A, Oliveira GMM, Correia LCL, Ramos Júnior AN, Luquetti AO, Hasslocher-Moreno AM, Sousa ASD, Paola AAVD, Sousa ACS, Ribeiro ALP, Correia Filho D, Souza DDSMD, Cunha-Neto E, Ramires FJA, Bacal F, Nunes MDCP, Martinelli Filho M, Scanavacca MI, Saraiva RM, Oliveira Júnior WAD, Lorga-Filho AM, Guimarães ADJBDA, Braga ALL, Oliveira ASD, Sarabanda AVL, Pinto AYDN, Carmo AALD, Schmidt A, Costa ARD, Ianni BM, Markman Filho B, Rochitte CE, Macêdo CT, Mady C, Chevillard C, Virgens CMBD, Castro CND, Britto CFDPDC, Pisani C, Rassi DDC, Sobral Filho DC, Almeida DRD, Bocchi EA, Mesquita ET, Mendes FDSNS, Gondim FTP, Silva GMSD, Peixoto GDL, Lima GGD, Veloso HH, Moreira HT, Lopes HB, Pinto IMF, Ferreira JMBB, Nunes JPS, Barreto-Filho JAS, Saraiva JFK, Lannes-Vieira J, Oliveira JLM, Armaganijan LV, Martins LC, Sangenis LHC, Barbosa MPT, Almeida-Santos MA, Simões MV, Yasuda MAS, Moreira MDCV, Higuchi MDL, Monteiro MRDCC, Mediano MFF, Lima MM, Oliveira MTD, Romano MMD, Araujo NNSLD, Medeiros PDTJ, Alves RV, Teixeira RA, Pedrosa RC, Aras Junior R, Torres RM, Povoa RMDS, Rassi SG, Alves SMM, Tavares SBDN, Palmeira SL, Silva Júnior TLD, Rodrigues TDR, Madrini Junior V, Brant VMDC, Dutra WO, Dias JCP. SBC Guideline on the Diagnosis and Treatment of Patients with Cardiomyopathy of Chagas Disease - 2023. Arq Bras Cardiol 2023; 120:e20230269. [PMID: 37377258 PMCID: PMC10344417 DOI: 10.36660/abc.20230269] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023] Open
Affiliation(s)
- José Antonio Marin-Neto
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | - Anis Rassi
- Hospital do Coração Anis Rassi , Goiânia , GO - Brasil
| | | | | | | | - Alejandro Ostermayer Luquetti
- Centro de Estudos da Doença de Chagas , Hospital das Clínicas da Universidade Federal de Goiás , Goiânia , GO - Brasil
| | | | - Andréa Silvestre de Sousa
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Antônio Carlos Sobral Sousa
- Universidade Federal de Sergipe , São Cristóvão , SE - Brasil
- Hospital São Lucas , Rede D`Or São Luiz , Aracaju , SE - Brasil
| | | | | | | | - Edecio Cunha-Neto
- Universidade de São Paulo , Faculdade de Medicina da Universidade, São Paulo , SP - Brasil
| | - Felix Jose Alvarez Ramires
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Fernando Bacal
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Martino Martinelli Filho
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Maurício Ibrahim Scanavacca
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Roberto Magalhães Saraiva
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Adalberto Menezes Lorga-Filho
- Instituto de Moléstias Cardiovasculares , São José do Rio Preto , SP - Brasil
- Hospital de Base de Rio Preto , São José do Rio Preto , SP - Brasil
| | | | | | - Adriana Sarmento de Oliveira
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Ana Yecê das Neves Pinto
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Andre Schmidt
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | - Andréa Rodrigues da Costa
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | - Barbara Maria Ianni
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Carlos Eduardo Rochitte
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
- Hcor , Associação Beneficente Síria , São Paulo , SP - Brasil
| | | | - Charles Mady
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Christophe Chevillard
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Marselha - França
| | | | | | | | - Cristiano Pisani
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | | | - Edimar Alcides Bocchi
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Evandro Tinoco Mesquita
- Hospital Universitário Antônio Pedro da Faculdade Federal Fluminense , Niterói , RJ - Brasil
| | | | | | | | | | | | - Henrique Horta Veloso
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | - Henrique Turin Moreira
- Hospital das Clínicas , Faculdade de Medicina de Ribeirão Preto , Universidade de São Paulo , Ribeirão Preto , SP - Brasil
| | | | | | | | - João Paulo Silva Nunes
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
- Fundação Zerbini, Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | | | | | | | - Luiz Cláudio Martins
- Universidade Estadual de Campinas , Faculdade de Ciências Médicas , Campinas , SP - Brasil
| | | | | | | | - Marcos Vinicius Simões
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | | | | | - Maria de Lourdes Higuchi
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Mauro Felippe Felix Mediano
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
- Instituto Nacional de Cardiologia (INC), Rio de Janeiro, RJ - Brasil
| | - Mayara Maia Lima
- Secretaria de Vigilância em Saúde , Ministério da Saúde , Brasília , DF - Brasil
| | | | | | | | | | - Renato Vieira Alves
- Instituto René Rachou , Fundação Oswaldo Cruz , Belo Horizonte , MG - Brasil
| | - Ricardo Alkmim Teixeira
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Roberto Coury Pedrosa
- Hospital Universitário Clementino Fraga Filho , Instituto do Coração Edson Saad - Universidade Federal do Rio de Janeiro , RJ - Brasil
| | | | | | | | | | - Silvia Marinho Martins Alves
- Ambulatório de Doença de Chagas e Insuficiência Cardíaca do Pronto Socorro Cardiológico Universitário da Universidade de Pernambuco (PROCAPE/UPE), Recife , PE - Brasil
| | | | - Swamy Lima Palmeira
- Secretaria de Vigilância em Saúde , Ministério da Saúde , Brasília , DF - Brasil
| | | | | | - Vagner Madrini Junior
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | - João Carlos Pinto Dias
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| |
Collapse
|
14
|
Altcheh J, Moscatelli G, Caruso M, Moroni S, Bisio M, Miranda MR, Monla C, Vaina M, Valdez M, Moran L, Ramirez T, Patiño OL, Riarte A, Gonzalez N, Fernandes J, Alves F, Ribeiro I, Garcia-Bournissen F. Population pharmacokinetics of benznidazole in neonates, infants and children using a new pediatric formulation. PLoS Negl Trop Dis 2023; 17:e0010850. [PMID: 37256863 DOI: 10.1371/journal.pntd.0010850] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/23/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND There is a major need for information on pharmacokinetics (PK) of benznidazole (BNZ) in children with Chagas disease (CD). We conducted a multicentre population PK, safety and efficacy study in children, infants and neonates with CD treated with BNZ (formulated in 100 mg tablets or 12.5 mg dispersible tablets, developed by the pharmaceutical company LAFEPE, in a collaboration with DNDi). METHODS 81 children 0-12 years old were enrolled at 5 pediatric centers in Argentina. Diagnosis of T. cruzi infection was confirmed by direct microscopic examination, or at least two positive conventional serological tests. Subject enrolment was stratified by age: newborns to 2 years (minimum of 10 newborns) and >2-12 years. BNZ 7.5 mg/kg/d was administered in two daily doses for 60 days. Five blood samples per child were obtained at random times within pre-defined time windows at Day 0 at 2-5 h post-dose; during steady state, one sample at Day 7 and at Day 30; and two samples at 12-24 h after final BNZ dose at Day 60. The primary efficacy endpoint was parasitological clearance by qualitative PCR at the end of treatment. RESULTS Forty-one (51%) patients were under 2 years of age (including 14 newborns <1 month of age). Median age at enrolment was 22 months (mean: 43.2; interquartile range (IQR) 7-72 months). The median measured BNZ Cmax was 8.32 mg/L (IQR 5.95-11.8; range 1.79-19.38). Median observed BNZ Cmin (trough) concentration was 2 mg/L (IQR 1.25-3.77; range 0.14-7.08). Overall median simulated Css was 6.3 mg/L (IQR 4.7-8.5 mg/L). CL/F increased quickly during the first month of postnatal life and reached adult levels after approximately 10 years of age. Negative qPCR was observed at the end of treatment in all 76 patients who completed the treatment. Five patients discontinued treatment (3 due to AEs and 2 due to lack of compliance). CONCLUSION We observed lower BNZ plasma concentrations in infants and children than those previously reported in adults treated with comparable mg/kg doses. Despite these lower concentrations, pediatric treatment was well tolerated and universally effective, with a high response rate and infrequent, mild AEs. TRIAL REGISTRATION Registered in clinicaltrials.gov #NCT01549236.
Collapse
Affiliation(s)
- Jaime Altcheh
- Servicio de Parasitologia y Chagas, Hospital de Niños "Dr Ricardo Gutierrez", Buenos Aires, Argentina
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Instituto de Investigaciones en Patologias Pediatricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Guillermo Moscatelli
- Servicio de Parasitologia y Chagas, Hospital de Niños "Dr Ricardo Gutierrez", Buenos Aires, Argentina
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Instituto de Investigaciones en Patologias Pediatricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Martin Caruso
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Hospital de Niños Doctor Hector Quintana, Jujuy, Argentina
| | - Samanta Moroni
- Servicio de Parasitologia y Chagas, Hospital de Niños "Dr Ricardo Gutierrez", Buenos Aires, Argentina
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Instituto de Investigaciones en Patologias Pediatricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Margarita Bisio
- Servicio de Parasitologia y Chagas, Hospital de Niños "Dr Ricardo Gutierrez", Buenos Aires, Argentina
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Instituto de Investigaciones en Patologias Pediatricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Maria Rosa Miranda
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Hospital de Niños Doctor Hector Quintana, Jujuy, Argentina
| | - Celia Monla
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Hospital Público Materno Infantil, Salta, Argentina
| | - Maria Vaina
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Hospital Público Materno Infantil, Salta, Argentina
| | - Maria Valdez
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Hospital Público Materno Infantil, Salta, Argentina
| | - Lucrecia Moran
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Centro de Chagas y Patología Regional, Santiago del Estero, Argentina
| | - Teresa Ramirez
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
| | - Oscar Ledesma Patiño
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Centro de Chagas y Patología Regional, Santiago del Estero, Argentina
| | - Adelina Riarte
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Instituto Nacional de Parasitología Dr. Mario Fatala Chaben, Buenos Aires, Argentina
| | - Nicolas Gonzalez
- Servicio de Parasitologia y Chagas, Hospital de Niños "Dr Ricardo Gutierrez", Buenos Aires, Argentina
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Instituto de Investigaciones en Patologias Pediatricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Jayme Fernandes
- Drugs for Neglected Diseases initiative, Rio de Janeiro, Brazil
| | - Fabiana Alves
- Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Isabela Ribeiro
- Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Facundo Garcia-Bournissen
- Servicio de Parasitologia y Chagas, Hospital de Niños "Dr Ricardo Gutierrez", Buenos Aires, Argentina
- PEDCHAGAS Network (Hospital de Niños Ricardo Gutiérrez, Hospital de Niños Doctor Hector Quintana, Hospital Público Materno Infantil, Centro de Chagas y Patología Regional de Santiago del Estero, & Instituto Nacional de Parasitología Dr. Mario Fatala Chaben), Argentina
- Division of Paediatric Clinical Pharmacology, Department of Paediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
15
|
García-Estrada C, Pérez-Pertejo Y, Domínguez-Asenjo B, Holanda VN, Murugesan S, Martínez-Valladares M, Balaña-Fouce R, Reguera RM. Further Investigations of Nitroheterocyclic Compounds as Potential Antikinetoplastid Drug Candidates. Biomolecules 2023; 13:biom13040637. [PMID: 37189384 DOI: 10.3390/biom13040637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Due to the lack of specific vaccines, management of the trypanosomatid-caused neglected tropical diseases (sleeping sickness, Chagas disease and leishmaniasis) relies exclusively on pharmacological treatments. Current drugs against them are scarce, old and exhibit disadvantages, such as adverse effects, parenteral administration, chemical instability and high costs which are often unaffordable for endemic low-income countries. Discoveries of new pharmacological entities for the treatment of these diseases are scarce, since most of the big pharmaceutical companies find this market unattractive. In order to fill the pipeline of compounds and replace existing ones, highly translatable drug screening platforms have been developed in the last two decades. Thousands of molecules have been tested, including nitroheterocyclic compounds, such as benznidazole and nifurtimox, which had already provided potent and effective effects against Chagas disease. More recently, fexinidazole has been added as a new drug against African trypanosomiasis. Despite the success of nitroheterocycles, they had been discarded from drug discovery campaigns due to their mutagenic potential, but now they represent a promising source of inspiration for oral drugs that can replace those currently on the market. The examples provided by the trypanocidal activity of fexinidazole and the promising efficacy of the derivative DNDi-0690 against leishmaniasis seem to open a new window of opportunity for these compounds that were discovered in the 1960s. In this review, we show the current uses of nitroheterocycles and the novel derived molecules that are being synthesized against these neglected diseases.
Collapse
Affiliation(s)
- Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Bárbara Domínguez-Asenjo
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Vanderlan Nogueira Holanda
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Pilani 333031, India
| | - María Martínez-Valladares
- Instituto de Ganadería de Montaña (IGM), Consejo Superior de Investigaciones Científicas-Universidad de León, Carretera León-Vega de Infanzones, Vega de Infanzones, 24346 León, Spain
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rosa M. Reguera
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| |
Collapse
|
16
|
Pandey RP, Nascimento MS, Franco CH, Bortoluci K, Silva MN, Zingales B, Gibaldi D, Castaño Barrios L, Lannes-Vieira J, Cariste LM, Vasconcelos JR, Moraes CB, Freitas-Junior LH, Kalil J, Alcântara L, Cunha-Neto E. Drug Repurposing in Chagas Disease: Chloroquine Potentiates Benznidazole Activity against Trypanosoma cruzi In Vitro and In Vivo. Antimicrob Agents Chemother 2022; 66:e0028422. [PMID: 36314800 PMCID: PMC9664849 DOI: 10.1128/aac.00284-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/16/2022] [Indexed: 11/20/2022] Open
Abstract
Drug combinations and drug repurposing have emerged as promising strategies to develop novel treatments for infectious diseases, including Chagas disease. In this study, we aimed to investigate whether the repurposed drugs chloroquine (CQ) and colchicine (COL), known to inhibit Trypanosoma cruzi infection in host cells, could boost the anti-T. cruzi effect of the trypanocidal drug benznidazole (BZN), increasing its therapeutic efficacy while reducing the dose needed to eradicate the parasite. The combination of BZN and COL exhibited cytotoxicity to infected cells and low antiparasitic activity. Conversely, a combination of BZN and CQ significantly reduced T. cruzi infection in vitro, with no apparent cytotoxicity. This effect seemed to be consistent across different cell lines and against both the partially BZN-resistant Y and the highly BZN-resistant Colombiana strains. In vivo experiments in an acute murine model showed that the BZN+CQ combination was eight times more effective in reducing T. cruzi infection in the acute phase than BZN monotherapy. In summary, our results demonstrate that the concomitant administration of CQ and BZN potentiates the trypanocidal activity of BZN, leading to a reduction in the dose needed to achieve an effective response. In a translational context, it could represent a higher efficacy of treatment while also mitigating the adverse effects of high doses of BZN. Our study also reinforces the relevance of drug combination and repurposing approaches in the field of Chagas disease drug discovery.
Collapse
Affiliation(s)
- Ramendra P. Pandey
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
- School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Marilda Savoia Nascimento
- School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute of Tropical Medicine of São Paulo, São Paulo, Brazil
| | - Caio Haddad Franco
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Karina Bortoluci
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil
| | - Marcelo Nunes Silva
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Bianca Zingales
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Leonardo Moro Cariste
- Recombinant Vaccines Laboratory, Department of Biosciences, Federal University of São Paulo, São Paulo, Brazil
| | - Jose Ronnie Vasconcelos
- Recombinant Vaccines Laboratory, Department of Biosciences, Federal University of São Paulo, São Paulo, Brazil
- Department of Microbiology, Immunology, and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Carolina Borsoi Moraes
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lucio H. Freitas-Junior
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jorge Kalil
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
- School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Laura Alcântara
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
- School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute of Tropical Medicine of São Paulo, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
- School of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Klein K, Roberti J, Rouvier M, Belizan M, Cafferata ML, Berrueta AM, Alonso JP. Design and feasibility of an implementation strategy to address Chagas guidelines engagement focused on attending women of childbearing age and children at the primary healthcare level in Argentina: a pilot study. BMC PRIMARY CARE 2022; 23:277. [PMID: 36348310 PMCID: PMC9643922 DOI: 10.1186/s12875-022-01886-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 10/22/2022] [Indexed: 11/11/2022]
Abstract
Background Chagas is a public health problem, especially in Latin America, exacerbated by migratory movements and increasing urbanization. Argentina is among the countries with the highest estimated prevalence in the region, with 1,500,000 people infected, with mother to child as the main mode of transmission. Vertical transmission has been significantly reduced by treating women of childbearing age; several guidelines in the region recommend treatment as a primary prevention strategy for the child and a secondary prevention strategy for women and their families. Despite recommendations, women of childbearing age are not always treated, and children do not receive timely diagnosis and treatment. The objective of this research was to design an implementation strategy to improve using Chagas guidelines focused on attending women of childbearing age and children at the primary healthcare level and pilot it in three primary health care centers in Argentina. Methods We conducted a pilot feasibility study using the Consolidated Framework for Implementation Research. A qualitative process evaluation was conducted using semi-structured interviews with health care providers and observations in primary health care centers. Results We developed a multifaceted implementation strategy including training, flowcharts and reminders, a register of suspected and confirmed cases, and the selection of a management facilitator. The pilot study took place between September 2019 and May 2020. The implementation level was heterogeneous and varied depending on the components, being the facilitating factors, the simplicity of the intervention, professionals’ willingness to expand the indication of serologic tests, and staff commitment to the adoption of intervention components. The main barriers found were the change of authorities at the local level, some professionals´ reluctance to administer etiological treatment, staff shortages, lack of diagnostic supplies, and the health emergency caused by the COVID-19 pandemic. Conclusions Behavioral change strategies should be applied to improve implementation to address some of the main barriers, including support actions offered by opinion leaders, medical experts, and local health authorities. Rapid diagnostic tests should be readily available to maintain behavior changes. We suggest further refinement of the strategy and its implementation in more centers to assess outcomes prospectively with a hybrid implementation research design. Supplementary Information The online version contains supplementary material available at 10.1186/s12875-022-01886-6.
Collapse
Affiliation(s)
- Karen Klein
- Institute of Clinical Effectiveness and Health Policy (IECS), City of Buenos Aires, Argentina.
| | - Javier Roberti
- Institute of Clinical Effectiveness and Health Policy (IECS), City of Buenos Aires, Argentina.,National Scientific and Technical Research Council (CONICET), City of Buenos Aires, Argentina
| | - Mariel Rouvier
- Ministry of Public Health of Chaco, Resistencia, Chaco, Argentina
| | - Maria Belizan
- Institute of Clinical Effectiveness and Health Policy (IECS), City of Buenos Aires, Argentina
| | - Maria Luisa Cafferata
- Institute of Clinical Effectiveness and Health Policy (IECS), City of Buenos Aires, Argentina
| | - Amanda Mabel Berrueta
- Institute of Clinical Effectiveness and Health Policy (IECS), City of Buenos Aires, Argentina
| | - Juan Pedro Alonso
- Institute of Clinical Effectiveness and Health Policy (IECS), City of Buenos Aires, Argentina.,National Scientific and Technical Research Council (CONICET), City of Buenos Aires, Argentina
| |
Collapse
|
18
|
Soeiro MDNC. Perspectives for a new drug candidate for Chagas disease therapy. Mem Inst Oswaldo Cruz 2022; 117:e220004. [PMID: 35293439 PMCID: PMC8923671 DOI: 10.1590/0074-02760220004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/17/2022] [Indexed: 12/21/2022] Open
Abstract
Chagas disease (CD), a neglected tropical illness caused by the protozoan Trypanosoma cruzi, affects more than 6 million people mostly in poor areas of Latin America. CD has two phases: an acute, short phase mainly oligosymptomatic followed to the chronic phase, a long-lasting stage that may trigger cardiac and/or digestive disorders and death. Only two old drugs are available and both present low efficacy in the chronic stage, display side effects and are inactive against parasite strains naturally resistant to these nitroderivatives. These shortcomings justify the search for novel therapeutic options considering the target product profile for CD that will be presently reviewed besides briefly revisiting the data on phosphodiesterase inhibitors upon T. cruzi.
Collapse
|
19
|
Lascano F, García Bournissen F, Altcheh J. Review of pharmacological options for the treatment of Chagas disease. Br J Clin Pharmacol 2022; 88:383-402. [PMID: 33314266 DOI: 10.1111/bcp.14700] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/09/2020] [Accepted: 12/02/2020] [Indexed: 12/20/2022] Open
Abstract
Chagas disease (CD) is a worldwide problem, with over 8 million people infected in both rural and urban areas. CD was first described over a century ago, but only two drugs are currently available for CD treatment: benznidazole (BZN) and nifurtimox (NF). Treating CD-infected patients, especially children and women of reproductive age, is vital in order to prevent long-term sequelae, such as heart and gastrointestinal dysfunction, but this aim is still far from being accomplished. Currently, the strongest data to support benefit-risk considerations come from trials in children. Treatment response biomarkers need further development as serology is being questioned as the best method to assess treatment response. This article is a narrative review on the pharmacology of drugs for CD, particularly BZN and NF. Data on drug biopharmaceutical characteristics, safety and efficacy of both drugs are summarized from a clinical perspective. Current data on alternative compounds under evaluation for CD treatment, and new possible treatment response biomarkers are also discussed. Early diagnosis and treatment of CD, especially in paediatric patients, is vital for an effective and safe use of the available drugs (i.e. BZN and NF). New biomarkers for CD are urgently needed for the diagnosis and evaluation of treatment efficacy, and to guide efforts from academia and pharmaceutical companies to accelerate the process of new drug development.
Collapse
Affiliation(s)
- Fernanda Lascano
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Gobierno de la Ciudad de la Nación Argentina, Buenos Aires, Argentina.,Servicio de Parasitología y Chagas, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Facundo García Bournissen
- Division of Pediatric Clinical Pharmacology, Department of Pediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada
| | - Jaime Altcheh
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Gobierno de la Ciudad de la Nación Argentina, Buenos Aires, Argentina.,Servicio de Parasitología y Chagas, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| |
Collapse
|
20
|
Strauss M, Lo Presti MS, Ramírez JC, Bazán PC, Velázquez López DA, Báez AL, Paglini PA, Schijman AG, Rivarola HW. Differential tissue distribution of discrete typing units after drug combination therapy in experimental Trypanosoma cruzi mixed infection. Parasitology 2021; 148:1595-1601. [PMID: 35060468 PMCID: PMC11010057 DOI: 10.1017/s0031182021001281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/15/2021] [Accepted: 07/12/2021] [Indexed: 11/06/2022]
Abstract
The aim of the present work was to evaluate the distribution of the different clones of the parasite prevailing after treatment with benznidazole (BZ) and clomipramine (CLO), in mice infected with Trypanosoma cruzi, Casibla isolate which consists of a mixture of two discrete typing units (DTUs). Albino Swiss mice were infected and treated with high and low concentrations of BZ (100 or 6.25 mg/kg), CLO (5 or 1.25 mg/kg), or the combination of both low doses (BZ6.25 + CLO1.25), during the acute phase of experimental infection. Treatment efficacy was evaluated by comparing parasitaemia, survival and tissular parasite presence. For DTUs genotyping, blood, skeletal and cardiac muscle samples were analysed by multiplex quantitative polymerase chain reaction. The combined treatment had similar outcomes to BZ6.25; BZ100 was the most effective treatment, but it failed to reach parasite clearance and produced greater histological alterations. Non-treated mice and the ones treated with monotherapies showed both DTUs while BZ6.25 + CLO1.25 treated mice showed only TcVI parasites in all the tissues studied. These findings suggest that the treatment may modify the distribution of infecting DTUs in host tissues. Coinfection with T. cruzi clones belonging to different DTUs reveals a complex scenario for the treatment of Chagas disease and search for new therapies.
Collapse
Affiliation(s)
- Mariana Strauss
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC-CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Santa Rosa 1085, X5000ESU-Córdoba, Argentina
| | - M. Silvina Lo Presti
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC-CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Santa Rosa 1085, X5000ESU-Córdoba, Argentina
| | - Juan C. Ramírez
- Laboratorio de Biología Molecular de la Enfermedad de Chagas (LaBMECh), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - P. Carolina Bazán
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC-CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Santa Rosa 1085, X5000ESU-Córdoba, Argentina
| | - Daniela A. Velázquez López
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC-CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Santa Rosa 1085, X5000ESU-Córdoba, Argentina
| | - Alejandra L. Báez
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC-CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Santa Rosa 1085, X5000ESU-Córdoba, Argentina
| | - Patricia A. Paglini
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC-CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Santa Rosa 1085, X5000ESU-Córdoba, Argentina
| | - Alejandro G. Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas (LaBMECh), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI-CONICET), Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | - Héctor W. Rivarola
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC-CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Santa Rosa 1085, X5000ESU-Córdoba, Argentina
| |
Collapse
|
21
|
Ciapponi A, Barreira F, Perelli L, Bardach A, Gascón J, Molina I, Morillo C, Prado N, Riarte A, Torrico F, Ribeiro I, Villar JC, Sosa-Estani S. Fixed vs adjusted-dose benznidazole for adults with chronic Chagas disease without cardiomyopathy: A systematic review and meta-analysis. PLoS Negl Trop Dis 2020; 14:e0008529. [PMID: 32804966 PMCID: PMC7451967 DOI: 10.1371/journal.pntd.0008529] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 08/27/2020] [Accepted: 06/26/2020] [Indexed: 12/22/2022] Open
Abstract
Chagas disease is a neglected disease that remains a public health threat, particularly in Latin America. The most important treatment options are nitroimidazole derivatives, such as nifurtimox and benznidazole (BZN). Some studies suggest that for adults seropositive to T. cruzi but without clinically evident chronic Chagas cardiomyopathy (CCC), a simple fixed-dose scheme of BZN could be equivalent to a weight-adjusted dose. We compared the efficacy and safety of a fixed dose of BZN with an adjusted dose for T. cruzi seropositive adults without CCC. We used the Cochrane methods, and reported according to the PRISMA statement. We included randomized controlled trials (RCTs) allocating participants to fixed and/or adjusted doses of BZN for T. cruzi seropositive adults without CCC. We searched (December 2019) Cochrane, MEDLINE, EMBASE, LILACS, Clinicaltrials.gov, and International Clinical Trials Registry Platform (ICTRP), and contacted Chagas experts. Selection, data extraction, and risk of bias assessment, using the Cochrane tool, were performed independently by pairs of reviewers. Discrepancies were solved by consensus within the team. Primary outcomes were parasite-related outcomes and efficacy or patient-related safety outcomes. We conducted a meta-analysis using RevMan 5.3 software and used GRADE summary of finding tables to present the certainty of evidence by outcome. We identified 655 records through our search strategy and 10 studies (four of them ongoing) met our inclusion criteria. We did not find any study directly comparing fixed vs adjusted doses of BZN, however, some outcomes allowed subgroup comparisons between fixed and adjusted doses of BZN against placebo. Moderate-certainty evidence suggests no important subgroup differences for positive PCR at one year and for three safety outcomes (drug discontinuation, peripheral neuropathy, and mild rash). The same effect was observed for any serious adverse events (low-certainty evidence). All subgroups showed similar effects (I2 0% for all these subgroup comparisons but 32% for peripheral neuropathy), supporting the equivalence of BZN schemes. We conclude that there is no direct evidence comparing fixed and adjusted doses of BZN. Based on low to very low certainty of evidence for critical clinical outcomes and moderate certainty of evidence for important outcomes, fixed and adjusted doses may be equivalent in terms of safety and efficacy. An individual patient data network meta-analysis could better address this issue.
Collapse
Affiliation(s)
- Agustín Ciapponi
- Centro Cochrane Argentino-Instituto de Efectividad Clínica y Sanitaria (IECS-CONICET), Buenos Aires, Argentina
- Centro de Investigaciones Epidemiológicas y Salud Pública (CIESP-IECS). CONICET, Buenos Aires, Argentina
| | - Fabiana Barreira
- Drugs for Neglected Diseases initiative (DNDi), Río de Janeiro, Brazil
| | - Lucas Perelli
- Centro Cochrane Argentino-Instituto de Efectividad Clínica y Sanitaria (IECS-CONICET), Buenos Aires, Argentina
| | - Ariel Bardach
- Centro Cochrane Argentino-Instituto de Efectividad Clínica y Sanitaria (IECS-CONICET), Buenos Aires, Argentina
- Centro de Investigaciones Epidemiológicas y Salud Pública (CIESP-IECS). CONICET, Buenos Aires, Argentina
| | | | - Israel Molina
- Hospital Universitari Vall d'Hebron Research Institute, Barcelona, España
| | - Carlos Morillo
- McMaster University, Population Health Research Institute, Hamilton, Canada
| | - Nilda Prado
- Instituto Nacional de Parasitología Dr. M Fatala Chaben, Buenos Aires, Argentina
| | - Adelina Riarte
- Instituto Nacional de Parasitología Dr. M Fatala Chaben, Buenos Aires, Argentina
| | | | - Isabela Ribeiro
- Drugs for Neglected Diseases initiative (DNDi), Geneva, Switzerland
| | - Juan Carlos Villar
- Grupo de Cardiología Preventiva, Facultad de Ciencias de la Salud, Universidad Autónoma de Bucaramanga, Bucaramanga, Colombia
| | - Sergio Sosa-Estani
- Centro de Investigaciones Epidemiológicas y Salud Pública (CIESP-IECS). CONICET, Buenos Aires, Argentina
- Drugs for Neglected Diseases initiative (DNDi), Río de Janeiro, Brazil
| |
Collapse
|