1
|
Razzaq S, Olmo F, Lakshminarayana SB, Ying-Bo C, Jayawardhana S, Rao SPS, Kelly JM, A. F. F. Statins do not reduce the parasite burden during experimental Trypanosoma cruzi infection. Antimicrob Agents Chemother 2025; 69:e0013525. [PMID: 40331859 PMCID: PMC12057342 DOI: 10.1128/aac.00135-25] [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: 01/30/2025] [Accepted: 03/09/2025] [Indexed: 05/08/2025] Open
Abstract
Cardiomyopathy is the most common pathology associated with Trypanosoma cruzi infection. Reports that statins have both cardioprotective and trypanocidal activity have generated interest in their potential as a therapeutic treatment. Using a highly sensitive bioluminescent mouse model, we show that a 5-day treatment with statins has no significant impact on parasite load. The free systemic concentrations fail to reach the level required for potency. Hence, clinical trials to investigate the trypanocidal activity of statins lack experimental justification.
Collapse
Affiliation(s)
- Sarah Razzaq
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Parasitology, Faculty of Sciences, University of Granada, Granada, Andalusia, Spain
| | | | - Chen Ying-Bo
- Global Health, Biomedical Research, Novartis, Emeryville, California, USA
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Francisco A. F.
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
2
|
Assmus F, Adehin A, Hoglund RM, Fortes Francisco A, Lewis MD, Kelly JM, Charman SA, White KL, Shackleford DM, Escudié F, Chatelain E, Scandale I, Tarning J. Pharmacokinetic-pharmacodynamic modeling of benznidazole and its antitrypanosomal activity in a murine model of chronic Chagas disease. PLoS Negl Trop Dis 2025; 19:e0012968. [PMID: 40359193 PMCID: PMC12074391 DOI: 10.1371/journal.pntd.0012968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/07/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND There is an urgent need for improved treatments for Chagas disease, a neglected tropical infection caused by the protozoan parasite Trypanosoma cruzi. Benznidazole, the first line therapy, has severe limitations such as poor tolerability and variable efficacy in the chronic stage of infection. To optimize dosing regimens, a better understanding of the pharmacokinetic/pharmacodynamic (PK/PD) relationship for benznidazole is crucial. This study aimed to characterize the population pharmacokinetic properties of benznidazole in mice and investigate the relationship between exposure and antitrypanosomal activity in T. cruzi infected mice. METHODOLOGY/PRINCIPAL FINDINGS Antitrypanosomal activity was assessed in 118 BALB/c mice with chronic-stage T. cruzi infection, utilizing highly sensitive in vivo bioluminescence imaging (BLI). Benznidazole was administered at doses ranging from 10 to 100 mg/kg for 5-20 days. The pharmacokinetic properties of benznidazole were evaluated in 52 uninfected BALB/c mice using nonlinear mixed-effects modeling. The relationship between simulated benznidazole exposure and sterile parasitological cure in the BLI experiments was evaluated by logistic regression and partial least squares - discriminant analysis. Benznidazole pharmacokinetics in mice were well described by a one-compartment disposition model with first-order absorption, with higher doses associated with slower absorption. Univariate logistic regression revealed a significant correlation between drug exposure and the probability of parasitological cure. Total plasma exposure, time above IC90 and peak plasma concentration were all strongly associated with efficacy, provided that benznidazole was administered for at least 5 days. CONCLUSIONS/SIGNIFICANCE This is the first study to successfully quantify the dose-response relationship for benznidazole in T. cruzi-infected mice using preclinical BLI data. Our results underscore the complexity of distinguishing PK/PD drivers of efficacy due to high collinearity between PK/PD index parameters, and we propose dose-fractionation studies for future research. Studying the PK/PD relationship using the BLI model provides valuable insights, aiding hypothesis generation through endpoint assessment of parasite infection.
Collapse
Affiliation(s)
- Frauke Assmus
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ayorinde Adehin
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Richard M. Hoglund
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Amanda Fortes Francisco
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Susan A. Charman
- Centre for Drug Candidate Optimisation, Monash University, Melbourne, Australia
| | - Karen L. White
- Centre for Drug Candidate Optimisation, Monash University, Melbourne, Australia
| | | | - Fanny Escudié
- Drugs for Neglected Disease initiative, Geneva, Switzerland
| | - Eric Chatelain
- Drugs for Neglected Disease initiative, Geneva, Switzerland
| | - Ivan Scandale
- Drugs for Neglected Disease initiative, Geneva, Switzerland
| | - Joel Tarning
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
3
|
de Oliveira R, Cruz LR, Dessoy MA, Koovits PJ, dos Santos DA, de Oliveira LFN, Ferreira RA, Mollo MC, Lee E, Duarte SM, Krogh R, Ferreira LLG, Chelucci RC, Dichiara M, Simpson QJ, Feltrin C, da Silva AC, dos Santos BM, Broering MF, Pollastri MP, Ferrins L, Moraes CB, Andricopulo AD, Kratz JM, Sjö P, Mowbray CE, Dias LC. Discovery and Early Optimization of 1 H-Indole-2-carboxamides with Anti- Trypanosoma cruzi Activity. J Med Chem 2025; 68:7313-7340. [PMID: 40163677 PMCID: PMC11998015 DOI: 10.1021/acs.jmedchem.4c02942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/08/2025] [Accepted: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Chagas disease (CD), caused by the flagellate protozoan Trypanosoma cruzi, is a neglected tropical disease endemic in 21 countries. The only two antiparasitic drugs approved for its treatment, benznidazole and nifurtimox, have significant drawbacks. We present herein the optimization of a series of substituted indoles that were identified through phenotypic screening against T. cruzi. Early lead compounds with balanced potency and physicochemical properties were advanced to animal studies but showed limited plasma exposure. Medicinal chemistry strategies were used to improve metabolic stability and solubility, but unfortunately, this effort failed to yield compounds with improvements in both exposure and potency. Still, the best compound was progressed for a proof-of-concept efficacy study using acute and chronic mice models of Chagas disease. Despite showing antiparasitic activity in these in vivo studies, the optimization work with this series was stopped due to unfavorable drug metabolism and pharmacokinetic (DMPK) properties and a deprioritized mechanism of action (CYP51 inhibition).
Collapse
Affiliation(s)
- Ramon
G. de Oliveira
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | - Luiza R. Cruz
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
- Drugs
for Neglected Diseases initiative, Rio de Janeiro 20010-020, Brazil
| | - Marco A. Dessoy
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | - Paul J. Koovits
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | | | | | - Rafael A. Ferreira
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | - María C. Mollo
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | - Eun Lee
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | - Simone M. Duarte
- São
Carlos Institute of Physics, University
of São Paulo, São
Carlos 13563-120, Brazil
| | - Renata Krogh
- São
Carlos Institute of Physics, University
of São Paulo, São
Carlos 13563-120, Brazil
| | | | - Rafael C. Chelucci
- São
Carlos Institute of Physics, University
of São Paulo, São
Carlos 13563-120, Brazil
| | - Maria Dichiara
- Chemistry and Chemical Biology and Pharmaceutical
Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Quillon J. Simpson
- Chemistry and Chemical Biology and Pharmaceutical
Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Clarissa Feltrin
- Institute of Biomedical Sciences and School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Adriana C. da Silva
- Institute of Biomedical Sciences and School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Benedito M. dos Santos
- Institute of Biomedical Sciences and School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Milena F. Broering
- Institute of Biomedical Sciences and School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Michael P. Pollastri
- Chemistry and Chemical Biology and Pharmaceutical
Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Lori Ferrins
- Chemistry and Chemical Biology and Pharmaceutical
Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Carolina B. Moraes
- Institute of Biomedical Sciences and School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Jadel M. Kratz
- Drugs
for Neglected Diseases initiative, Rio de Janeiro 20010-020, Brazil
| | - Peter Sjö
- Drugs
for Neglected Diseases initiative, Geneva 1202, Switzerland
| | | | - Luiz C. Dias
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| |
Collapse
|
4
|
Khan AA, Taylor MC, Fortes Francisco A, Jayawardhana S, Atherton RL, Olmo F, Lewis MD, Kelly JM. Animal models for exploring Chagas disease pathogenesis and supporting drug discovery. Clin Microbiol Rev 2024; 37:e0015523. [PMID: 39545730 PMCID: PMC11629624 DOI: 10.1128/cmr.00155-23] [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] [Indexed: 11/17/2024] Open
Abstract
SUMMARYInfections with the parasitic protozoan Trypanosoma cruzi cause Chagas disease, which results in serious cardiac and/or digestive pathology in 30%-40% of individuals. However, symptomatic disease can take decades to become apparent, and there is a broad spectrum of possible outcomes. The complex and long-term nature of this infection places a major constraint on the scope for experimental studies in humans. Accordingly, predictive animal models have been a mainstay of Chagas disease research. The resulting data have made major contributions to our understanding of parasite biology, immune responses, and disease pathogenesis and have provided a platform that informs and facilitates the global drug discovery effort. Here, we provide an overview of available animal models and illustrate how they have had a key impact across the field.
Collapse
Affiliation(s)
- Archie A. Khan
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Amanda Fortes Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Richard L. Atherton
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
5
|
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
|
6
|
González S, Wall RJ, Thomas J, Braillard S, Brunori G, Díaz IC, Cantizani J, Carvalho S, Castañeda Casado P, Chatelain E, Cotillo I, Fiandor JM, Francisco AF, Grimsditch D, Keenan M, Kelly JM, Kessler A, Luise C, Lyon JJ, MacLean L, Marco M, Martin JJ, Martinez MS, Paterson C, Read KD, Santos-Villarejo A, Zuccotto F, Wyllie S, Miles TJ, De Rycker M. Short-course combination treatment for experimental chronic Chagas disease. Sci Transl Med 2023; 15:eadg8105. [PMID: 38091410 PMCID: PMC7615676 DOI: 10.1126/scitranslmed.adg8105] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023]
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, affects millions of people in the Americas and across the world, leading to considerable morbidity and mortality. Current treatment options, benznidazole (BNZ) and nifurtimox, offer limited efficacy and often lead to adverse side effects because of long treatment durations. Better treatment options are therefore urgently required. Here, we describe a pyrrolopyrimidine series, identified through phenotypic screening, that offers an opportunity to improve on current treatments. In vitro cell-based washout assays demonstrate that compounds in the series are incapable of killing all parasites; however, combining these pyrrolopyrimidines with a subefficacious dose of BNZ can clear all parasites in vitro after 5 days. These findings were replicated in a clinically predictive in vivo model of chronic Chagas disease, where 5 days of treatment with the combination was sufficient to prevent parasite relapse. Comprehensive mechanism of action studies, supported by ligand-structure modeling, show that compounds from this pyrrolopyrimidine series inhibit the Qi active site of T. cruzi cytochrome b, part of the cytochrome bc1 complex of the electron transport chain. Knowledge of the molecular target enabled a cascade of assays to be assembled to evaluate selectivity over the human cytochrome b homolog. As a result, a highly selective and efficacious lead compound was identified. The combination of our lead compound with BNZ rapidly clears T. cruzi parasites, both in vitro and in vivo, and shows great potential to overcome key issues associated with currently available treatments.
Collapse
Affiliation(s)
- Silvia González
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Richard J. Wall
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - John Thomas
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | | | | | - Juan Cantizani
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Sandra Carvalho
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | | | - Ignacio Cotillo
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Jose M. Fiandor
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | | | | | | | - John M. Kelly
- London School for Hygiene and Tropical Medicine, London, UK
| | - Albane Kessler
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Chiara Luise
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | - Lorna MacLean
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Maria Marco
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - J. Julio Martin
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | | | - Christy Paterson
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Kevin D. Read
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | - Fabio Zuccotto
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Susan Wyllie
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Tim J. Miles
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Manu De Rycker
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| |
Collapse
|
7
|
Milagre MM, Torchelsen FKVDS, Pedrosa TCF, Teixeira GM, Sampaio LS, Saúde-Guimarães DA, Branquinho RT, Mosqueira VCF, Lana MD. Lychnopholide loaded in surface modified polylactide nanocapsules (LYC-PLA-PEG-NC) cure mice infected by Trypanosoma cruzi strain a prototype of resistance to benznidazole and nifurtimox: First insights of its mechanism of action. Exp Parasitol 2023; 255:108647. [PMID: 37914151 DOI: 10.1016/j.exppara.2023.108647] [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: 04/04/2023] [Revised: 09/21/2023] [Accepted: 10/28/2023] [Indexed: 11/03/2023]
Abstract
Chagas disease (CD) remains neglected and causes high morbidity and mortality. The great difficulty is the lack of effective treatment. The current drugs cause side effects and have limited therapeutic efficacy in the chronic phase. This study aims to fulfil some gaps in studies of the natural substance lychnopholide nanoencapsulated LYC-PLA-PEG-NC (LYC-NC) and free (Free-LYC): the activity in epimastigotes and amastigotes to determine its selectivity index (SI), the therapeutic efficacy in mice infected with Colombian Trypanosoma cruzi strain and insight of the mechanism of LYC-NC action on T. cruzi. The SI was obtained by calculation of the ratio between the IC50 value toward H9c2 cells divided by the IC50 value in the anti-T. cruzi test. Infected Swiss mice were treated with 2 and 12 mg/kg/day via intravenous and oral, respectively, and the therapeutic efficacy was determined. The IC50 of LYC-NC and Free-LYC for epimastigotes of T. cruzi were similar. Both were active against amastigotes in cell culture, particularly Free-LYC. The SI of LYC-NC and Free-LYC were 45.38 and 32.11, respectively. LYC-NC 2 and 12 mg/kg/day cured parasitologically, 62.5% and 80% of the animals, respectively, infected with a strain resistant to treatment. The fluorescent NC was distributed in the cardiomyocyte cytoplasm, infected or not, and interacted with the trypomastigotes. Together, these results represent advances in demonstrating LYC as a potent new therapeutic option for treating CD.
Collapse
Affiliation(s)
- Matheus Marques Milagre
- Universidade Federal de Ouro Preto, Escola de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | | | | | - Gabriel Marques Teixeira
- Universidade Federal de Ouro Preto, Escola de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Larissa Silva Sampaio
- Universidade Federal de Ouro Preto, Escola de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Dênia Antunes Saúde-Guimarães
- Universidade Federal de Ouro Preto, Escola de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Renata Tupinambá Branquinho
- Universidade Federal de Ouro Preto, Escola de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Vanessa Carla Furtado Mosqueira
- Universidade Federal de Ouro Preto, Escola de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Marta de Lana
- Universidade Federal de Ouro Preto, Escola de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Ouro Preto, Minas Gerais, 35400-000, Brazil.
| |
Collapse
|
8
|
Dean DA, Roach J, Ulrich vonBargen R, Xiong Y, Kane SS, Klechka L, Wheeler K, Jimenez Sandoval M, Lesani M, Hossain E, Katemauswa M, Schaefer M, Harris M, Barron S, Liu Z, Pan C, McCall LI. Persistent Biofluid Small-Molecule Alterations Induced by Trypanosoma cruzi Infection Are Not Restored by Parasite Elimination. ACS Infect Dis 2023; 9:2173-2189. [PMID: 37883691 PMCID: PMC10842590 DOI: 10.1021/acsinfecdis.3c00261] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Chagas disease (CD), caused by Trypanosoma cruzi (T. cruzi) protozoa, is a complicated parasitic illness with inadequate medical measures for diagnosing infection and monitoring treatment success. To address this gap, we analyzed changes in the metabolome of T. cruzi-infected mice via liquid chromatography tandem mass spectrometry of clinically accessible biofluids: saliva, urine, and plasma. Urine was the most indicative of infection status across mouse and parasite genotypes. Metabolites perturbed by infection in urine include kynurenate, acylcarnitines, and threonylcarbamoyladenosine. Based on these results, we sought to implement urine as a tool for the assessment of CD treatment success. Strikingly, it was found that mice with parasite clearance following benznidazole antiparasitic treatment had an overall urine metabolome comparable to that of mice that failed to clear parasites. These results provide a complementary hypothesis to explain clinical trial data in which benznidazole treatment did not improve patient outcomes in late-stage disease, even in patients with successful parasite clearance. Overall, this study provides insights into new small-molecule-based CD diagnostic methods and a new approach to assess functional responses to treatment.
Collapse
Affiliation(s)
- Danya A. Dean
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
| | - Jarrod Roach
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | | | - Yi Xiong
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Shelley S. Kane
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
| | - London Klechka
- Department of Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Kate Wheeler
- Department of Biology, University of Oklahoma, Norman, OK, 73019, USA
| | | | - Mahbobeh Lesani
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Ekram Hossain
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
| | - Mitchelle Katemauswa
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
| | - Miranda Schaefer
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Morgan Harris
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Sayre Barron
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Zongyuan Liu
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
| | - Chongle Pan
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| |
Collapse
|
9
|
Jayawardhana S, Ward AI, Francisco AF, Lewis MD, Taylor MC, Kelly JM, Olmo F. Benznidazole treatment leads to DNA damage in Trypanosoma cruzi and the persistence of rare widely dispersed non-replicative amastigotes in mice. PLoS Pathog 2023; 19:e1011627. [PMID: 37956215 PMCID: PMC10681306 DOI: 10.1371/journal.ppat.1011627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/27/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Benznidazole is the front-line drug used to treat infections with Trypanosoma cruzi, the causative agent of Chagas disease. However, for reasons that are unknown, treatment failures are common. When we examined parasites that survived benznidazole treatment in mice using highly sensitive in vivo and ex vivo bioluminescence imaging, we found that recrudescence is not due to persistence of parasites in a specific organ or tissue that preferentially protects them from drug activity. Surviving parasites are widely distributed and located in host cells where the vast majority contained only one or two amastigotes. Therefore, infection relapse does not arise from a small number of intact large nests. Rather, persisters are either survivors of intracellular populations where co-located parasites have been killed, or amastigotes in single/low-level infected cells exist in a state where they are less susceptible to benznidazole. To better assess the nature of parasite persisters, we exposed infected mammalian cell monolayers to a benznidazole regimen that reduces the intracellular amastigote population to <1% of the pre-treatment level. Of host cells that remained infected, as with the situation in vivo, the vast majority contained only one or two surviving intracellular amastigotes. Analysis, based on non-incorporation of the thymidine analogue EdU, revealed these surviving parasites to be in a transient non-replicative state. Furthermore, treatment with benznidazole led to widespread parasite DNA damage. When the small number of parasites which survive in mice after non-curative treatment were assessed using EdU labelling, this revealed that these persisters were also initially non-replicative. A possible explanation could be that triggering of the T. cruzi DNA damage response pathway by the activity of benznidazole metabolites results in exit from the cell cycle as parasites attempt DNA repair, and that metabolic changes associated with non-proliferation act to reduce drug susceptibility. Alternatively, a small percentage of the parasite population may pre-exist in this non-replicative state prior to treatment.
Collapse
Affiliation(s)
- Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Alexander I. Ward
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Amanda F. Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
10
|
Francisco AF, Chen G, Wang W, Sykes ML, Escudié F, Scandale I, Olmo F, Shackleford DM, Zulfiqar B, Kratz JM, Pham T, Saunders J, Hu M, Avery VM, Charman SA, Kelly JM, Chatelain E. Preclinical data do not support the use of amiodarone or dronedarone as antiparasitic drugs for Chagas disease at the approved human dosing regimen. FRONTIERS IN TROPICAL DISEASES 2023; 4. [DOI: 10.3389/fitd.2023.1254061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
The repurposing of approved drugs is an appealing method to fast-track the development of novel therapies for neglected diseases. Amiodarone and dronedarone, two approved antiarrhythmic agents, have been reported to have potential for the management of Chagas disease patients displaying symptomatic heart pathology. More recently, it has been suggested that both molecules not only have an antiarrhythmic effect, but also have trypanocidal activity against Trypanosoma cruzi, the causative agent of Chagas disease. In this work, we assessed the in vitro activity of these compounds against T. cruzi, the in vivo pharmacokinetics, and pharmacodynamics, to determine the potential for repurposing these drugs as therapies for Chagas disease. Based on these results, we were unable to reproduce the in vitro potencies of amiodarone and dronedarone described in the literature, and both drugs were found to be inactive or cytotoxic against a variety of different mammalian cell lines. The evaluation of in vivo efficacy in a bioluminescent murine model of T. cruzi did not show antiparasitic activity at the highest tolerated dose tested. While the potential of amiodarone and dronedarone as antiarrhythmic agents in Chagas cardiomyopathic patients cannot be completely excluded, a trypanocidal effect in patients treated with these two drugs appears unlikely.
Collapse
|
11
|
Dean DA, Roach J, vonBargen RU, Xiong Y, Kane SS, Klechka L, Wheeler K, Sandoval MJ, Lesani M, Hossain E, Katemauswa M, Schaefer M, Harris M, Barron S, Liu Z, Pan C, McCall LI. Persistent biofluid small molecule alterations induced by Trypanosoma cruzi infection are not restored by antiparasitic treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.03.543565. [PMID: 37425694 PMCID: PMC10326868 DOI: 10.1101/2023.06.03.543565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Chagas Disease (CD), caused by Trypanosoma cruzi (T. cruzi) protozoa, is a complicated parasitic illness with inadequate medical measures for diagnosing infection and monitoring treatment success. To address this gap, we analyzed changes in the metabolome of T. cruzi-infected mice via liquid chromatography tandem mass spectrometry analysis of clinically-accessible biofluids: saliva, urine, and plasma. Urine was the most indicative of infection status, across mouse and parasite genotypes. Metabolites perturbed by infection in the urine include kynurenate, acylcarnitines, and threonylcarbamoyladenosine. Based on these results, we sought to implement urine as a tool for assessment of CD treatment success. Strikingly, it was found that mice with parasite clearance following benznidazole antiparasitic treatment had comparable overall urine metabolome to mice that failed to clear parasites. These results match with clinical trial data in which benznidazole treatment did not improve patient outcomes in late-stage disease. Overall, this study provides insights into new small molecule-based CD diagnostic methods and a new approach to assess functional treatment response.
Collapse
Affiliation(s)
- Danya A. Dean
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019; USA
| | - Jarrod Roach
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | | | - Yi Xiong
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Shelley S. Kane
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019; USA
| | - London Klechka
- Department of Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Kate Wheeler
- Department of Biology, University of Oklahoma, Norman, OK, 73019, USA
| | | | - Mahbobeh Lesani
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Ekram Hossain
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019; USA
| | - Mitchelle Katemauswa
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019; USA
| | - Miranda Schaefer
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Morgan Harris
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Sayre Barron
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Zongyuan Liu
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019; USA
| | - Chongle Pan
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, 73019; USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, 73019, USA
| |
Collapse
|
12
|
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
|
13
|
Tarleton RL. Effective drug discovery in Chagas disease. Trends Parasitol 2023; 39:423-431. [PMID: 37024318 DOI: 10.1016/j.pt.2023.03.015] [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: 02/24/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 04/07/2023]
Abstract
The Chagas field has gone >50 years without tangible progress toward new therapies. My colleagues and I have recently reported on a benzoxaborole compound that achieves consistent parasitological cure in experimentally infected mice and in naturally infected non-human primates (NHPs). While these results do not assure success in human clinical trials, they significantly de-risk this process and form a strong justification for such trials. Highly effective drug discovery depends on a solid understanding of host and parasite biology and excellent knowledge in designing and validating chemical entities. This opinion piece seeks to provide perspectives on the process that led to the discovery of AN15368, with the hope that this will facilitate the discovery of additional clinical candidates for Chagas disease.
Collapse
Affiliation(s)
- Rick L Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
14
|
Tayama Y, Mizukami S, Toume K, Komatsu K, Yanagi T, Nara T, Tieu P, Huy NT, Hamano S, Hirayama K. Anti-Trypanosoma cruzi activity of Coptis rhizome extract and its constituents. Trop Med Health 2023; 51:12. [PMID: 36859380 PMCID: PMC9976467 DOI: 10.1186/s41182-023-00502-2] [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: 12/30/2022] [Accepted: 02/09/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Current therapeutic agents, including nifurtimox and benznidazole, are not sufficiently effective in the chronic phase of Trypanosoma cruzi infection and are accompanied by various side effects. In this study, 120 kinds of extracts from medicinal herbs used for Kampo formulations and 94 kinds of compounds isolated from medicinal herbs for Kampo formulations were screened for anti-T. cruzi activity in vitro and in vivo. METHODS As an experimental method, a recombinant protozoan cloned strain expressing luciferase, namely Luc2-Tulahuen, was used in the experiments. The in vitro anti-T. cruzi activity on epimastigote, trypomastigote, and amastigote forms was assessed by measuring luminescence intensity after treatment with the Kampo extracts or compounds. In addition, the cytotoxicity of compounds was tested using mouse and human feeder cell lines. The in vivo anti-T. cruzi activity was measured by a murine acute infection model using intraperitoneal injection of trypomastigotes followed by live bioluminescence imaging. RESULTS As a result, three protoberberine-type alkaloids, namely coptisine chloride, dehydrocorydaline nitrate, and palmatine chloride, showed strong anti-T. cruzi activities with low cytotoxicity. The IC50 values of these compounds differed depending on the side chain, and the most effective compound, coptisine chloride, showed a significant effect in the acute infection model. CONCLUSIONS For these reasons, coptisine chloride is a hit compound that can be a potential candidate for anti-Chagas disease drugs. In addition, it was expected that there would be room for further improvement by modifying the side chains of the basic skeleton.
Collapse
Affiliation(s)
- Yuki Tayama
- grid.174567.60000 0000 8902 2273Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ,grid.174567.60000 0000 8902 2273Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan
| | - Shusaku Mizukami
- grid.174567.60000 0000 8902 2273Department of Immune Regulation, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan ,grid.174567.60000 0000 8902 2273School of Tropical Medicines and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ,grid.174567.60000 0000 8902 2273The Joint Usage/Research Center On Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523 Japan
| | - Kazufumi Toume
- grid.267346.20000 0001 2171 836XSection of Pharmacognosy, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Katsuko Komatsu
- grid.267346.20000 0001 2171 836XSection of Pharmacognosy, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Tetsuo Yanagi
- grid.174567.60000 0000 8902 2273NEKKEN Bio-Resource Center (NBRC), Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan ,grid.174567.60000 0000 8902 2273The Joint Usage/Research Center On Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523 Japan
| | - Takeshi Nara
- grid.411789.20000 0004 0371 1051Faculty of Pharmacy, Iryo Sosei University, Iwaki, Fukushima Japan
| | - Paul Tieu
- grid.25073.330000 0004 1936 8227Faculty of Health Sciences, McMaster University, Hamilton, ON Canada ,Online Research Club, Nagasaki, Japan
| | - Nguyen Tien Huy
- grid.174567.60000 0000 8902 2273Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ,grid.174567.60000 0000 8902 2273School of Tropical Medicines and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ,Online Research Club, Nagasaki, Japan
| | - Shinjiro Hamano
- grid.174567.60000 0000 8902 2273Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan ,grid.174567.60000 0000 8902 2273Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan ,grid.174567.60000 0000 8902 2273The Joint Usage/Research Center On Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523 Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan. .,Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan. .,School of Tropical Medicines and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan. .,The Joint Usage/Research Center On Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523, Japan.
| |
Collapse
|
15
|
Ciapponi A, Barreira F, Perelli L, Bardach A, Gascón J, Molina I, Morillo C, Prado N, Riarte A, Torrico F, Villar JC, Reidel S, Gibbons L, Sosa-Estani S. Direct evidence gap on fixed versus adjusted-dose benznidazole for adults with chronic Chagas disease without cardiomyopathy: Systematic review and individual patient data meta-analysis. Trop Med Int Health 2023; 28:2-16. [PMID: 36420767 DOI: 10.1111/tmi.13831] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES To determine the comparative efficacy and safety of a fixed dose of benznidazole (BZN) with an adjusted-dose for Trypanosoma cruzi-seropositive adults without cardiomyopathy. METHODS We conducted a systematic review and individual participant data (IPD) meta-analysis following Cochrane methods, and the PRISMA-IPD statement for reporting. Randomised controlled trials (RCTs) allocating participants to fixed or adjusted doses of BZN for T. cruzi-seropositive adults without cardiomyopathy were included. We searched (December 2021) Cochrane, MEDLINE, EMBASE, LILACS and trial registries and contacted Chagas experts. Selection, data extraction, risk of bias assessment using the Cochrane tool, and a GRADE summary of finding tables were performed independently by pairs of reviewers. We conducted a random-effects IPD meta-analysis using the one-stage strategy, or, if that was impossible, the two-stage strategy. RESULTS Five RCTs (1198 patients) were included, none directly comparing fixed with adjusted doses of BZN. Compared to placebo, BZN therapy was strongly associated with negative qPCR and sustainable parasitological clearance regardless of the type of dose and subgroup analysed. For negative qPCR, the fixed/adjusted rate of odds ratios (RORF/A ) was 8.83 (95% CI 1.02-76.48); for sustained parasitological clearance, it was 4.60 (95% CI 0.40-52.51), probably indicating at least non-inferior effect of fixed doses, with no statistically significant interactions by scheme for global and most subgroup estimations. The RORF/A for treatment interruption due to adverse events was 0.44 (95% CI 0.14-1.38), probably indicating no worse tolerance of fixed doses. CONCLUSIONS We found no direct comparison between fixed and adjusted doses of BZN. However, fixed doses versus placebo are probably not inferior to weight-adjusted doses of BZN versus placebo in terms of parasitological efficacy and safety. Network IPD meta-analysis, through indirect comparisons, may well provide the best possible answers in the near future. REGISTRATION The study protocol was registered in PROSPERO (CRD42019120905).
Collapse
Affiliation(s)
- Agustín Ciapponi
- Departamento de Evaluación de Tecnologías Sanitarias, Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina.,Centro de Investigaciones Epidemiológicas y Salud Pública, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Fabiana Barreira
- Chagas Clinical Project, Drugs for Neglected Diseases Initiative, Río de Janeiro, Brazil
| | - Lucas Perelli
- Departamento de Evaluación de Tecnologías Sanitarias, Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | - Ariel Bardach
- Departamento de Evaluación de Tecnologías Sanitarias, Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina.,Centro de Investigaciones Epidemiológicas y Salud Pública, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Joaquim Gascón
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública, Madrid, Spain
| | - Israel Molina
- Infectious Diseases Department, Hospital Universitario Vall d'Hebron, Barcelona, Spain.,International Health Program, Catalan Institute of Health, Barcelona, Spain
| | - Carlos Morillo
- Libin Cardiovascular Institute, University of Calgary, Calgary, Canada
| | - Nilda Prado
- Instituto Nacional de Parasitología Dr. M Fatala Chaben, Ministerio de Salud de la Nación, Buenos Aires, Argentina
| | - Adelina Riarte
- Instituto Nacional de Parasitología Dr. M Fatala Chaben, Ministerio de Salud de la Nación, Buenos Aires, Argentina
| | - Faustino Torrico
- Parasitología y Enfermedades Infecciosas, Universidad Mayor de San Simón, Cochabamba, Bolivia
| | - Juan Carlos Villar
- Departamento de Investigaciones, Fundación Cardioinfantil, Instituto de Cardiología, Bogotá, Colombia
| | - Sara Reidel
- Departamento de Evaluación de Tecnologías Sanitarias, Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | - Luz Gibbons
- Departamento de Evaluación de Tecnologías Sanitarias, Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | - Sergio Sosa-Estani
- Centro de Investigaciones Epidemiológicas y Salud Pública, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Chagas Clinical Project, Drugs for Neglected Diseases Initiative, Río de Janeiro, Brazil
| |
Collapse
|
16
|
Identification of Aryl Polyamines Derivatives as Anti- Trypanosoma cruzi Agents Targeting Iron Superoxide Dismutase. Pharmaceutics 2022; 15:pharmaceutics15010140. [PMID: 36678771 PMCID: PMC9863987 DOI: 10.3390/pharmaceutics15010140] [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: 11/03/2022] [Revised: 12/13/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023] Open
Abstract
Chagas disease (CD) is a tropical and potentially fatal infection caused by Trypanosoma cruzi. Although CD was limited to Latin America as a silent disease, CD has become widespread as a result of globalization. Currently, 6-8 million people are infected worldwide, and no effective treatment is available. Here, we identify new effective agents against T. cruzi. In short, 16 aryl polyamines were screened in vitro against different T. cruzi strains, and lead compounds were evaluated in vivo after oral administration in both the acute and chronic infections. The mode of action was also evaluated at the energetic level, and its high activity profile could be ascribed to a mitochondria-dependent bioenergetic collapse and redox stress by inhibition of the Fe-SOD enzyme. We present compound 15 as a potential compound that provides a step forward for the development of new agents to combat CD.
Collapse
|
17
|
Zuma AA, de Souza W. Fexinidazole interferes with the growth and structural organization of Trypanosoma cruzi. Sci Rep 2022; 12:20388. [PMID: 36437273 PMCID: PMC9701812 DOI: 10.1038/s41598-022-23941-z] [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: 07/11/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022] Open
Abstract
Fexinidazole (FEX) is a heterocyclic compound and constitutes the first 100% oral treatment drug for African trypanosomiasis. Its effectiveness against Trypanosoma brucei encouraged the investigation of its antiparasitic potential against T. cruzi, the aetiological agent of Chagas disease. Although previous studies addressed the antitrypanosomal effects of FEX, none used electron microscopy to identify the main target structures of T. brucei or T. cruzi. In this work, we used microscopy techniques to analyze the ultrastructural alterations caused by FEX in different developmental stages of T. cruzi. In addition to inhibiting T. cruzi proliferation, with IC50 of 1 µM for intracellular amastigotes, FEX promoted massive disorganization of reservosomes, the detachment of the plasma membrane, unpacking of nuclear heterochromatin, mitochondrial swelling, Golgi disruption and alterations in the kinetoplast-mitochondrion complex. Together, these observations point to FEX as a potential drug leader for further developing of chemotherapy against Chagas disease.
Collapse
Affiliation(s)
- Aline Araujo Zuma
- grid.8536.80000 0001 2294 473XLaboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Centro de Ciências da Saúde, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ 21491-590 Brazil
| | - Wanderley de Souza
- grid.8536.80000 0001 2294 473XLaboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Centro de Ciências da Saúde, Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ 21491-590 Brazil ,grid.412290.c0000 0000 8024 0602Centro Multidisciplinar de Pesquisas Biológica-CMABio, Escola Superior de Ciências da Saúde, Universidade do Estado do Amazonas-UEA, Av. Carvalho Leal, 1777-Cachoeirinha, Manaus, AM 69065-000 Brazil
| |
Collapse
|
18
|
Francisco AF, Saade U, Jayawardhana S, Pottel H, Scandale I, Chatelain E, Liehl P, Kelly JM, Zrein M. Comparing in vivo bioluminescence imaging and the Multi-Cruzi immunoassay platform to develop improved Chagas disease diagnostic procedures and biomarkers for monitoring parasitological cure. PLoS Negl Trop Dis 2022; 16:e0010827. [PMID: 36190992 PMCID: PMC9560623 DOI: 10.1371/journal.pntd.0010827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/13/2022] [Accepted: 09/16/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Chagas disease is caused by the protozoan parasite Trypanosoma cruzi and is a serious public health problem throughout Latin America. With 6 million people infected, there is a major international effort to develop new drugs. In the chronic phase of the disease, the parasite burden is extremely low, infections are highly focal at a tissue/organ level, and bloodstream parasites are only intermittently detectable. As a result, clinical trials are constrained by difficulties associated with determining parasitological cure. Even highly sensitive PCR methodologies can be unreliable, with a tendency to produce "false-cure" readouts. Improved diagnostic techniques and biomarkers for cure are therefore an important medical need. METHODOLOGY/PRINCIPAL FINDINGS Using an experimental mouse model, we have combined a multiplex assay system and highly sensitive bioluminescence imaging to evaluate serological procedures for diagnosis of T. cruzi infections and confirmation of parasitological cure. We identified a set of three antigens that in the context of the multiplex serology system, provide a rapid, reactive and highly accurate read-out of both acute and chronic T. cruzi infection. In addition, we describe specific antibody responses where down-regulation can be correlated with benznidazole-mediated parasite reduction and others where upregulation is associated with persistent infection. One specific antibody (IBAG39) highly correlated with the bioluminescence flux and represents a promising therapy monitoring biomarker in mice. CONCLUSIONS/SIGNIFICANCE Robust, high-throughput methodologies for monitoring the efficacy of anti-T. cruzi drug treatment are urgently required. Using our experimental systems, we have identified markers of infection or parasite reduction that merit assessing in a clinical setting for the longitudinal monitoring of drug-treated patients.
Collapse
Affiliation(s)
- Amanda Fortes Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
| | | | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
| | - Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Ivan Scandale
- Drugs for Neglected Diseases initiative (DNDi), Geneva, Switzerland
| | - Eric Chatelain
- Drugs for Neglected Diseases initiative (DNDi), Geneva, Switzerland
| | | | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
- * E-mail:
| | | |
Collapse
|
19
|
Ang CW, Lee BM, Jackson CJ, Wang Y, Franzblau SG, Francisco AF, Kelly JM, Bernhardt PV, Tan L, West NP, Sykes ML, Hinton AO, Bolisetti R, Avery VM, Cooper MA, Blaskovich MA. Nitroimidazopyrazinones with Oral Activity against Tuberculosis and Chagas Disease in Mouse Models of Infection. J Med Chem 2022; 65:13125-13142. [DOI: 10.1021/acs.jmedchem.2c00972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Chee Wei Ang
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
- School of Science, Monash University Malaysia, Subang Jaya, 47500 Selangor, Malaysia
| | - Brendon M. Lee
- Research School of Chemistry, Australian National University, Sullivans Creek Road, Acton ACT 2601, Australia
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medical College, New York, New York 10021, United States
| | - Colin J. Jackson
- Research School of Chemistry, Australian National University, Sullivans Creek Road, Acton ACT 2601, Australia
| | - Yuehong Wang
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Scott G. Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Amanda F. Francisco
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Paul V. Bernhardt
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Lendl Tan
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas P. West
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Melissa L. Sykes
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
| | - Alexandra O. Hinton
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Raghu Bolisetti
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Vicky M. Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| | - Matthew A. Cooper
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Mark A.T. Blaskovich
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
20
|
Martín-Escolano R, Rosales MJ, Marín C. Biological characteristics of the Trypanosoma cruzi Arequipa strain make it a good model for Chagas disease drug discovery. Acta Trop 2022; 236:106679. [PMID: 36096184 DOI: 10.1016/j.actatropica.2022.106679] [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: 05/15/2022] [Revised: 09/04/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022]
Abstract
Trypanosoma cruzi, the causative agent of Chagas disease (CD), is a genuine parasite with tremendous genetic diversity and a complex life cycle. Scientists have studied this disease for more than 100 years, and CD drug discovery has been a mainstay due to the absence of an effective treatment. Technical advances in several areas have contributed to a better understanding of the complex biology and life cycle of this parasite, with the aim of designing the ideal profile of both drug and therapeutic options to treat CD. Here, we present the T. cruzi Arequipa strain (MHOM/Pe/2011/Arequipa) as an interesting model for CD drug discovery. We characterized acute-phase parasitaemia and chronic-phase tropism in BALB/c mice and determined the in vitro and in vivo benznidazole susceptibility profile of the different morphological forms of this strain. The tropism of this strain makes it an interesting model for the screening of new compounds with a potential anti-Chagas profile for the treatment of this disease.
Collapse
Affiliation(s)
- Rubén Martín-Escolano
- Laboratory of Molecular & Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK.
| | - María José Rosales
- Department of Parasitology, University of Granada, Severo Ochoa s/n, Granada 18071, Spain
| | - Clotilde Marín
- Department of Parasitology, University of Granada, Severo Ochoa s/n, Granada 18071, Spain.
| |
Collapse
|
21
|
Kratz JM, Gonçalves KR, Romera LM, Moraes CB, Bittencourt-Cunha P, Schenkman S, Chatelain E, Sosa-Estani S. The translational challenge in Chagas disease drug development. Mem Inst Oswaldo Cruz 2022; 117:e200501. [PMID: 35613156 PMCID: PMC9128742 DOI: 10.1590/0074-02760200501] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
Chagas disease is a neglected tropical disease caused by the protozoan parasite Trypanosoma cruzi. There is an urgent need for safe, effective, and accessible new treatments since the currently approved drugs have serious limitations. Drug development for Chagas disease has historically been hampered by the complexity of the disease, critical knowledge gaps, and lack of coordinated R&D efforts. This review covers some of the translational challenges associated with the progression of new chemical entities from preclinical to clinical phases of development, and discusses how recent technological advances might allow the research community to answer key questions relevant to the disease and to overcome hurdles in R&D for Chagas disease.
Collapse
Affiliation(s)
- Jadel M Kratz
- Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Karolina R Gonçalves
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brasil
| | - Lavínia Md Romera
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brasil
| | - Carolina Borsoi Moraes
- Universidade Federal de São Paulo, Departamento de Ciências Farmacêuticas, Diadema, SP, Brasil
| | - Paula Bittencourt-Cunha
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brasil.,Universidade Federal de São Paulo, Departamento de Microbiologia, Imunologia e Parasitologia, São Paulo, SP, Brasil
| | - Sergio Schenkman
- Universidade Federal de São Paulo, Departamento de Microbiologia, Imunologia e Parasitologia, São Paulo, SP, Brasil
| | - Eric Chatelain
- Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Sergio Sosa-Estani
- Drugs for Neglected Diseases initiative, Geneva, Switzerland.,Epidemiology and Public Health Research Centre, CIESP-CONICET, Buenos Aires, Argentina
| |
Collapse
|
22
|
Assessment of a combined treatment with a therapeutic vaccine and benznidazole for the Trypanosoma cruzi chronic infection. Acta Trop 2022; 229:106334. [PMID: 35101415 DOI: 10.1016/j.actatropica.2022.106334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 02/07/2023]
Abstract
The difficulties encountered in achieving treatments for chronic Chagas disease have promoted the investigation of new therapeutic strategies. In this study, we used two murine models of Trypanosoma cruzi chronic infection to determine the usefulness of applying a therapeutic vaccine alone or followed by benznidazole (Bz) chemotherapy. A vaccine formulation based on an N-terminal fragment of Trans-sialidase (TS) and Immunostimulant Particle Adjuvant (ISPA) - TSNt-ISPA was obtained. Firstly, the immunogenicity and protective capacity of TSNt-ISPA was demonstrated as a prophylactic formulation in an acute model of infection. Later, the formulation was assessed as a therapeutic vaccine alone or combined with (Bz) using two models of chronic infection. BALB/c mice chronically infected with Sylvio X10/4 or Tulahuen cl2 T. cruzi strains were not treated as control or treated only with the therapeutic vaccine TSNt-ISPA, with a combined treatment TSNt-ISPA+Bz (Bz applied after the vaccine), or only with Bz. The vaccination schedule consisted of TSNt-ISPA administration at days110, 120, and 130 post-infection (pi) and Bz administration was performed daily from day 140 to 170 pi. At day 273 pi, electrocardiographic (ECG) parameters, heart parasite load, myocarditis, and heart fibrosis were assessed. In both models, therapeutic administration of TSNt-ISPA reduced ECG alterations and the cardiac tissue damage observed in the chronic phase. Moreover, vaccine treatment significantly decreased heart parasite load in both Sylvio X10/4 and Tulahuen cl2 infected mice. The combined treatment, but not Bz or vaccine administration alone, allowed to restore ECG parameters in Tulahuen cl2 infected mice. The results indicate the usefulness of the therapeutic TSNt-ISPA formulation in BALB/c mice chronically infected with Sylvio X10/4 or Tulahuen cl2 strain. For the mice infected with T. cruzi Tulahuen cl2 strain, the combined treatment with the vaccine and Bz had a more positive effect on the course of heart disease than the individual treatments with the vaccine or Bz alone.
Collapse
|
23
|
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
|
24
|
Cantizani J, Gamallo P, Cotillo I, Alvarez-Velilla R, Martin J. Rate-of-Kill (RoK) assays to triage large compound sets for Chagas disease drug discovery: Application to GSK Chagas Box. PLoS Negl Trop Dis 2021; 15:e0009602. [PMID: 34270544 PMCID: PMC8318231 DOI: 10.1371/journal.pntd.0009602] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/28/2021] [Accepted: 06/28/2021] [Indexed: 11/18/2022] Open
Abstract
Chagas disease (CD) is a human disease caused by Trypanosoma cruzi. Whilst endemic in Latin America, the disease is spread around the world due to migration flows, being estimated that 8 million people are infected worldwide and over 10,000 people die yearly of complications linked to CD. Current chemotherapeutics is restricted to only two drugs, i.e. benznidazole (BNZ) and nifurtimox (NIF), both being nitroaromatic compounds sharing mechanism of action and exerting suboptimal efficacy and serious adverse effects. Recent clinical trials conducted to reposition antifungal azoles have turned out disappointing due to poor efficacy outcomes despite their promising preclinical profile. This apparent lack of translation from bench models to the clinic raises the question of whether we are using the right in vitro tools for compound selection. We propose that speed of action and cidality, rather than potency, are properties that can differentiate those compounds with better prospect of success to show efficacy in animal models of CD. Here we investigate the use of in vitro assays looking at the kinetics of parasite kill as a valuable surrogate to tell apart slow- (i.e. azoles targeting CYP51) and fast-acting (i.e. nitroaromatic) compounds. Data analysis and experimental design have been optimised to make it amenable for high-throughput compound profiling. Automated data reduction of experimental kinetic points to tabulated curve descriptors in conjunction with PCA, k-means and hierarchical clustering provide drug discoverers with a roadmap to guide navigation from hit qualification of a screening campaign to compound optimisation programs and assessment of combo therapy potential. As an example, we have studied compounds belonging to the GSK Chagas Box stemmed from the HTS campaign run against the full GSK 1.8 million compounds collection [1]. One of the challenges in early drug discovery of small molecules is the improvement of the poor success rate in the translation from in vitro biological profile into efficacy in disease models, and ultimately in the clinic. Reductionist in vitro models on the bench may not properly recapitulate disease biology, thus overlooking critical properties of candidate compounds. Chagas Disease is a neglected tropical disease caused by Trypanosoma cruzi, a protozoan parasite with a complex life cycle. Despite the promising prospect based on in vitro and in vivo preclinical studies, efforts to reposition antifungal azoles turned out to be disappointing in clinical trials, with treatment failure in Chagas patients. This raises the question of whether we are using the right preclinical tools for decision-making about moving compounds forward for the treatment of this disease. We hypothesise that in vitro potency and efficacy values alone might be distorting the translational power of preclinical compounds, and we propose the use of rate-of-kill (RoK) assays in high-throughput mode. Herewith we disclose a simple, systematic, and automated methodology of analysis of the otherwise complex kinetic patterns, which provides drug discoverers with a navigation guide along a compound optimisation program or prioritisation of best exemplars across different chemical series.
Collapse
Affiliation(s)
- Juan Cantizani
- Kinetoplastid DPU, Global Health R&D, GSK, Tres Cantos, Madrid, Spain
| | - Pablo Gamallo
- Kinetoplastid DPU, Global Health R&D, GSK, Tres Cantos, Madrid, Spain
| | - Ignacio Cotillo
- Kinetoplastid DPU, Global Health R&D, GSK, Tres Cantos, Madrid, Spain
| | | | - Julio Martin
- Kinetoplastid DPU, Global Health R&D, GSK, Tres Cantos, Madrid, Spain
- * E-mail:
| |
Collapse
|
25
|
Farani PSG, Begum K, Vilar-Pereira G, Pereira IR, Almeida IC, Roy S, Lannes-Vieira J, Moreira OC. Treatment With Suboptimal Dose of Benznidazole Mitigates Immune Response Molecular Pathways in Mice With Chronic Chagas Cardiomyopathy. Front Cell Infect Microbiol 2021; 11:692655. [PMID: 34381739 PMCID: PMC8351877 DOI: 10.3389/fcimb.2021.692655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/25/2021] [Indexed: 12/21/2022] Open
Abstract
Chronic Chagas cardiomyopathy (CCC) is the most frequent and severe form of Chagas disease, a neglected tropical illness caused by the protozoan Trypanosoma cruzi, and the main cause of morbimortality from cardiovascular problems in endemic areas. Although efforts have been made to understand the signaling pathways and molecular mechanisms underlying CCC, the immunological signaling pathways regulated by the etiological treatment with benznidazole (Bz) has not been reported. In experimental CCC, Bz combined with the hemorheological and immunoregulatory agent pentoxifylline (PTX) has beneficial effects on CCC. To explore the molecular mechanisms of Bz or Bz+PTX therapeutic strategies, C57BL/6 mice chronically infected with the T. cruzi Colombian strain (discrete typing unit TcI) and showing electrocardiographic abnormalities were submitted to suboptimal dose of Bz or Bz+PTX from 120 to 150 days postinfection. Electrocardiographic alterations, such as prolonged corrected QT interval and heart parasite load, were beneficially impacted by Bz and Bz+PTX. RT-qPCR TaqMan array was used to evaluate the expression of 92 genes related to the immune response in RNA extracted from heart tissues. In comparison with non-infected mice, 30 genes were upregulated, and 31 were downregulated in infected mice. Particularly, infection upregulated the cytokines IFN-γ, IL-12b, and IL-2 (126-, 44-, and 18-fold change, respectively) and the T-cell chemoattractants CCL3 and CCL5 (23- and 16-fold change, respectively). Bz therapy restored the expression of genes related to inflammatory response, cellular development, growth, and proliferation, and tissue development pathways, most probably linked to the cardiac remodeling processes inherent to CCC, thus mitigating the Th1-driven response found in vehicle-treated infected mice. The combined Bz+PTX therapy revealed pathways related to the modulation of cell death and survival, and organismal survival, supporting that this strategy may mitigate the progression of CCC. Altogether, our results contribute to the better understanding of the molecular mechanisms of the immune response in the heart tissue in chronic Chagas disease and reinforce that parasite persistence and dysregulated immune response underpin CCC severity. Therefore, Bz and Bz+PTX chemotherapies emerge as tools to interfere in these pathways aiming to improve CCC prognosis.
Collapse
Affiliation(s)
- Priscila Silva Grijó Farani
- Real Time PCR Platform RPT09A, Laboratory of Molecular Biology and Endemic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Khodeza Begum
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, United States
| | - Glaucia Vilar-Pereira
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Isabela Resende Pereira
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Igor C Almeida
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, United States
| | - Sourav Roy
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, United States
| | - Joseli Lannes-Vieira
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Otacilio Cruz Moreira
- Real Time PCR Platform RPT09A, Laboratory of Molecular Biology and Endemic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Taylor MC, Ward AI, Olmo F, Francisco AF, Jayawardhana S, Costa FC, Lewis MD, Kelly JM. Bioluminescent:Fluorescent Trypanosoma cruzi Reporter Strains as Tools for Exploring Chagas Disease Pathogenesis and Drug Activity. Curr Pharm Des 2021; 27:1733-1740. [PMID: 33234096 DOI: 10.2174/1381612826666201124113214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/16/2020] [Accepted: 07/23/2020] [Indexed: 11/22/2022]
Abstract
Chagas disease results from infection with the trypanosomatid parasite Trypanosoma cruzi. Progress in developing new drugs has been hampered by the long term and complex nature of the condition and by our limited understanding of parasite biology. Technical difficulties in assessing the parasite burden during the chronic stage of infection have also proven to be a particular challenge. In this context, the development of noninvasive, highly sensitive bioluminescence imaging procedures based on parasites that express a red-shifted luciferase has greatly enhanced our ability to monitor infections in experimental models. Applications of this methodology have led to new insights into tissue tropism and infection dynamics and have been a major driver in drug development. The system has been further modified by the generation of parasite reporter lines that express bioluminescent:fluorescent fusion proteins, an advancement that has allowed chronic infections in mice to be examined at a cellular level. By exploiting bioluminescence, to identify the rare sites of tissue infection, and fluorescence to detect T. cruzi at the level of individual host cells in histological sections, it has been possible to investigate the replication and differentiation status of parasites in vivo and to examine the cellular environment of infection foci. In combination, these data provide a framework for the detailed dissection of disease pathogenesis and drug activity.
Collapse
Affiliation(s)
- Martin C Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Alexander I Ward
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Amanda F Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Fernanda C Costa
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Michael D Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - John M Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| |
Collapse
|
27
|
Scarim CB, Olmo F, Ferreira EI, Chin CM, Kelly JM, Fortes Francisco A. Image-Based In Vitro Screening Reveals the Trypanostatic Activity of Hydroxymethylnitrofurazone against Trypanosoma cruzi. Int J Mol Sci 2021; 22:ijms22136930. [PMID: 34203228 PMCID: PMC8268475 DOI: 10.3390/ijms22136930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/14/2021] [Accepted: 06/23/2021] [Indexed: 12/23/2022] Open
Abstract
Hydroxymethylnitrofurazone (NFOH) is a therapeutic candidate for Chagas disease (CD). It has negligible hepatotoxicity in a murine model compared to the front-line drug benznidazole (BZN). Here, using Trypanosoma cruzi strains that express bioluminescent and/or fluorescent reporter proteins, we further investigated the in vitro and in vivo activity of NFOH to define whether the compound is trypanocidal or trypanostatic. The in vitro activity was assessed by exploiting the fluorescent reporter strain using wash-out assays and real-time microscopy. For animal experimentation, BALB/c mice were inoculated with the bioluminescent reporter strain and assessed by highly sensitive in vivo and ex vivo imaging. Cyclophosphamide treatment was used to promote parasite relapse in the chronic stage of infection. Our data show that NFOH acts by a trypanostatic mechanism, and that it is more active than BZN in vitro against the infectious trypomastigote form of Trypanosoma cruzi. We also found that it is more effective at curing experimental infections in the chronic stage, compared with the acute stage, a feature that it shares with BZN. Therefore, given its reduced toxicity, enhanced anti-trypomastigote activity, and curative properties, NFOH can be considered as a potential therapeutic option for Chagas disease, perhaps in combination with other trypanocidal agents.
Collapse
Affiliation(s)
- Cauê Benito Scarim
- School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, Sao Paulo 14800-903, Brazil; (C.B.S.); (C.M.C.)
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK; (F.O.); (J.M.K.)
| | - Francisco Olmo
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK; (F.O.); (J.M.K.)
| | - Elizabeth Igne Ferreira
- LAPEN—Laboratory of Design and Synthesis of Chemotherapeutic Agents Potentially Active on Neglected Diseases, Department of Pharmacy, School of Pharmaceutical Sciences, University of Sao Paulo (USP), Sao Paulo 05508-9000, Brazil;
| | - Chung Man Chin
- School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, Sao Paulo 14800-903, Brazil; (C.B.S.); (C.M.C.)
- Advanced Research Center in Medicine, School of Medicine, Union of the Colleges of the Great Lakes (UNILAGO), São José do Rio Preto, Sao Paulo 15030-070, Brazil
| | - John M. Kelly
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK; (F.O.); (J.M.K.)
| | - Amanda Fortes Francisco
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK; (F.O.); (J.M.K.)
- Correspondence: ; Tel.: +44-207-612-7864
| |
Collapse
|
28
|
Martín-Escolano R, Cebrián R, Maqueda M, Romero D, Rosales MJ, Sánchez-Moreno M, Marín C. Assessing the effectiveness of AS-48 in experimental mice models of Chagas' disease. J Antimicrob Chemother 2021; 75:1537-1545. [PMID: 32129856 DOI: 10.1093/jac/dkaa030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES We report the in vivo trypanocidal activity of the bacteriocin AS-48 (lacking toxicity), which is produced by Enterococcus faecalis, against the flagellated protozoan Trypanosoma cruzi, the aetiological agent of Chagas' disease. METHODS We determined the in vivo activity of AS-48 against the T. cruzi Arequipa strain in BALB/c mice (in both acute and chronic phases of Chagas' disease). We evaluated the parasitaemia, the reactivation of parasitaemia after immunosuppression and the nested parasites in the chronic phase by PCR in target tissues. RESULTS AS-48 reduced the parasitaemia profile in acute infection and showed a noteworthy reduction in the parasitic load in chronic infection after immunosuppression according to the results obtained by PCR (double-checking to demonstrate cure). CONCLUSIONS AS-48 is a promising alternative that provides a step forward in the development of a new therapy against Chagas' disease.
Collapse
Affiliation(s)
- Rubén Martín-Escolano
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, E-18071 Granada, Spain
| | - Rubén Cebrián
- Department of Microbiology, Faculty of Sciences, Avda. Fuentenueva s/n, University of Granada, 18071 Granada, Spain
| | - Mercedes Maqueda
- Department of Microbiology, Faculty of Sciences, Avda. Fuentenueva s/n, University of Granada, 18071 Granada, Spain
| | - Desirée Romero
- Department of Statistics and Operations Research, Faculty of Sciences, Avda. Fuentenueva s/n, University of Granada, 18071 Granada, Spain
| | - Maria José Rosales
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, E-18071 Granada, Spain
| | - Manuel Sánchez-Moreno
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, E-18071 Granada, Spain
| | - Clotilde Marín
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, E-18071 Granada, Spain
| |
Collapse
|
29
|
Martín-Escolano R, Etxebeste-Mitxeltorena M, Martín-Escolano J, Plano D, Rosales MJ, Espuelas S, Moreno E, Sánchez-Moreno M, Sanmartín C, Marín C. Selenium Derivatives as Promising Therapy for Chagas Disease: In Vitro and In Vivo Studies. ACS Infect Dis 2021; 7:1727-1738. [PMID: 33871252 PMCID: PMC8480776 DOI: 10.1021/acsinfecdis.1c00048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chagas disease is a tropical infection caused by the protozoan parasite Trypanosoma cruzi and a global public health concern. It is a paradigmatic example of a chronic disease without an effective treatment. Current treatments targeting T. cruzi are limited to two obsolete nitroheterocyclic drugs, benznidazole and nifurtimox, which lead to serious drawbacks. Hence, new, more effective, safer, and affordable drugs are urgently needed. Selenium and their derivatives have emerged as an interesting strategy for the treatment of different prozotoan diseases, such as African trypanosomiasis, leishmaniasis, and malaria. In the case of Chagas disease, diverse selenium scaffolds have been reported with antichagasic activity in vitro and in vivo. On the basis of these premises, we describe the in vitro and in vivo trypanocidal activity of 41 selenocompounds against the three morphological forms of different T. cruzi strains. For the most active selenocompounds, their effect on the metabolic and mitochondrial levels and superoxide dismutase enzyme inhibition capacity were measured in order to determine the possible mechanism of action. Derivative 26, with a selenocyanate motif, fulfills the most stringent in vitro requirements for potential antichagasic agents and exhibits a better profile than benznidazole in vivo. This finding provides a step forward for the development of a new antichagasic agent.
Collapse
Affiliation(s)
- Rubén Martín-Escolano
- Laboratory of Molecular & Evolutionary Parasitology, RAPID group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, United Kingdom
| | - Mikel Etxebeste-Mitxeltorena
- Facultad de Farmacia y Nutrición, Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea, 1, E-31008 Pamplona, Spain
- Instituto de Salud Tropical, Universidad de Navarra (ISTUN), Irunlarrea, 1, E-31008 Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Irunlarrea, 1, E-31008 Pamplona, Spain
| | - Javier Martín-Escolano
- Servicio de Microbiologia Clinica y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
- Instituto de Investigación, Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain
| | - Daniel Plano
- Facultad de Farmacia y Nutrición, Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea, 1, E-31008 Pamplona, Spain
- Instituto de Salud Tropical, Universidad de Navarra (ISTUN), Irunlarrea, 1, E-31008 Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Irunlarrea, 1, E-31008 Pamplona, Spain
| | - María J. Rosales
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs. Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | - Socorro Espuelas
- Facultad de Farmacia y Nutrición, Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea, 1, E-31008 Pamplona, Spain
- Instituto de Salud Tropical, Universidad de Navarra (ISTUN), Irunlarrea, 1, E-31008 Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Irunlarrea, 1, E-31008 Pamplona, Spain
| | - Esther Moreno
- Facultad de Farmacia y Nutrición, Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea, 1, E-31008 Pamplona, Spain
- Instituto de Salud Tropical, Universidad de Navarra (ISTUN), Irunlarrea, 1, E-31008 Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Irunlarrea, 1, E-31008 Pamplona, Spain
| | - Manuel Sánchez-Moreno
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs. Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | - Carmen Sanmartín
- Facultad de Farmacia y Nutrición, Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea, 1, E-31008 Pamplona, Spain
- Instituto de Salud Tropical, Universidad de Navarra (ISTUN), Irunlarrea, 1, E-31008 Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Irunlarrea, 1, E-31008 Pamplona, Spain
| | - Clotilde Marín
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs. Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| |
Collapse
|
30
|
Fonseca KDS, Perin L, de Paiva NCN, da Silva BC, Duarte THC, Marques FDS, Costa GDP, Molina I, Correa-Oliveira R, Vieira PMDA, Carneiro CM. Benznidazole Treatment: Time- and Dose-Dependence Varies with the Trypanosoma cruzi Strain. Pathogens 2021; 10:729. [PMID: 34207764 PMCID: PMC8229751 DOI: 10.3390/pathogens10060729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 12/02/2022] Open
Abstract
As the development of new drugs for Chagas disease is not a priority due to its neglected disease status, an option for increasing treatment adherence is to explore alternative treatment regimens, which may decrease the incidence of side effects. Therefore, we evaluated the efficacy of different therapeutic schemes with benznidazole (BNZ) on the acute and chronic phases of the disease, using mice infected with strains that have different BNZ susceptibilities. Our results show that the groups of animals infected by VL-10 strain, when treated in the chronic phase with a lower dose of BNZ for a longer period of time (40 mg/kg/day for 40 days) presented better treatment efficacy than with the standard protocol (100 mg/kg/day for 20 days) although the best result in the treatment of the animals infected by the VL-10 strain was with100 mg/kg/day for 40 days. In the acute infection by the Y and VL-10 strains of T. cruzi, the treatment with a standard dose, but with a longer time of treatment (100 mg/kg/day for 40 days) presented the best results. Given these data, our results indicate that for BNZ, the theory of dose and time proportionality does not apply to the phases of infection.
Collapse
Affiliation(s)
- Kátia da Silva Fonseca
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (K.d.S.F.); (L.P.); (N.C.N.d.P.); (B.C.d.S.); (G.d.P.C.); (I.M.); (R.C.-O.); (P.M.d.A.V.)
| | - Luísa Perin
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (K.d.S.F.); (L.P.); (N.C.N.d.P.); (B.C.d.S.); (G.d.P.C.); (I.M.); (R.C.-O.); (P.M.d.A.V.)
| | - Nívia Carolina Nogueira de Paiva
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (K.d.S.F.); (L.P.); (N.C.N.d.P.); (B.C.d.S.); (G.d.P.C.); (I.M.); (R.C.-O.); (P.M.d.A.V.)
| | - Beatriz Cristiane da Silva
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (K.d.S.F.); (L.P.); (N.C.N.d.P.); (B.C.d.S.); (G.d.P.C.); (I.M.); (R.C.-O.); (P.M.d.A.V.)
| | - Thays Helena Chaves Duarte
- Laboratory of Morphopathology, Department of Biological Sciences, Nucleus of Biological Sciences Research, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (T.H.C.D.); (F.d.S.M.)
| | - Flávia de Souza Marques
- Laboratory of Morphopathology, Department of Biological Sciences, Nucleus of Biological Sciences Research, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (T.H.C.D.); (F.d.S.M.)
| | - Guilherme de Paula Costa
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (K.d.S.F.); (L.P.); (N.C.N.d.P.); (B.C.d.S.); (G.d.P.C.); (I.M.); (R.C.-O.); (P.M.d.A.V.)
| | - Israel Molina
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (K.d.S.F.); (L.P.); (N.C.N.d.P.); (B.C.d.S.); (G.d.P.C.); (I.M.); (R.C.-O.); (P.M.d.A.V.)
- Tropical Medicine and International Health Unit, Department of Infectious Diseases, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, PROSICS Barcelona, 08035 Barcelona, Spain
| | - Rodrigo Correa-Oliveira
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (K.d.S.F.); (L.P.); (N.C.N.d.P.); (B.C.d.S.); (G.d.P.C.); (I.M.); (R.C.-O.); (P.M.d.A.V.)
- Laboratory of Cellular and Molecular Immunology, René Rachou Research Center, Oswaldo Cruz Foundation, Belo Horizonte 30190-002, Brazil
| | - Paula Melo de Abreu Vieira
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (K.d.S.F.); (L.P.); (N.C.N.d.P.); (B.C.d.S.); (G.d.P.C.); (I.M.); (R.C.-O.); (P.M.d.A.V.)
- Laboratory of Morphopathology, Department of Biological Sciences, Nucleus of Biological Sciences Research, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (T.H.C.D.); (F.d.S.M.)
| | - Cláudia Martins Carneiro
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil; (K.d.S.F.); (L.P.); (N.C.N.d.P.); (B.C.d.S.); (G.d.P.C.); (I.M.); (R.C.-O.); (P.M.d.A.V.)
- Department of Clinical Analysis, School of Pharmacy, Federal University of Ouro Preto, Ouro Preto 35400-000, Brazil
| |
Collapse
|
31
|
Martín-Escolano R, Molina-Carreño D, Plano D, Espuelas S, Rosales MJ, Moreno E, Aydillo C, Sanmartín C, Sánchez-Moreno M, Marín C. Library of Selenocyanate and Diselenide Derivatives as In Vivo Antichagasic Compounds Targeting Trypanosoma cruzi Mitochondrion. Pharmaceuticals (Basel) 2021; 14:ph14050419. [PMID: 34062791 PMCID: PMC8147293 DOI: 10.3390/ph14050419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Chagas disease is usually caused by tropical infection with the insect-transmitted protozoan Trypanosoma cruzi. Currently, Chagas disease is a major public health concern worldwide due to globalization, and there are no treatments neither vaccines because of the long-term nature of the disease and its complex pathology. Current treatments are limited to two obsolete drugs, benznidazole and nifurtimox, which lead to serious drawbacks. Taking into account the urgent need for strict research efforts to find new therapies, here, we describe the in vitro and in vivo trypanocidal activity of a library of selected forty-eight selenocyanate and diselenide derivatives that exhibited leishmanicidal properties. The inclusion of selenium, an essential trace element, was due to the well-known extensive pharmacological activities for selenium compounds including parasitic diseases as T. cruzi. Here we present compound 8 as a potential compound that exhibits a better profile than benznidazole both in vitro and in vivo. It shows a fast-acting behaviour that could be attributed to its mode of action: it acts in a mitochondrion-dependent manner, causing cell death by bioenergetic collapse. This finding provides a step forward for the development of a new antichagasic agent.
Collapse
Affiliation(s)
- Rubén Martín-Escolano
- Laboratory of Molecular & Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
- Correspondence: (R.M.-E.); (C.M.)
| | - Daniel Molina-Carreño
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios de Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain; (D.M.-C.); (M.J.R.); (M.S.-M.)
| | - Daniel Plano
- Facultad de Farmacia y Nutrición, Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea, E-31008 Pamplona, Spain; (D.P.); (S.E.); (E.M.); (C.A.); (C.S.)
- Instituto de Salud Tropical, Universidad de Navarra, ISTUN, Irunlarrea, E-31008 Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA) Irunlarrea, E-31008 Pamplona, Spain
| | - Socorro Espuelas
- Facultad de Farmacia y Nutrición, Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea, E-31008 Pamplona, Spain; (D.P.); (S.E.); (E.M.); (C.A.); (C.S.)
- Instituto de Salud Tropical, Universidad de Navarra, ISTUN, Irunlarrea, E-31008 Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA) Irunlarrea, E-31008 Pamplona, Spain
| | - María J. Rosales
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios de Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain; (D.M.-C.); (M.J.R.); (M.S.-M.)
| | - Esther Moreno
- Facultad de Farmacia y Nutrición, Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea, E-31008 Pamplona, Spain; (D.P.); (S.E.); (E.M.); (C.A.); (C.S.)
- Instituto de Salud Tropical, Universidad de Navarra, ISTUN, Irunlarrea, E-31008 Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA) Irunlarrea, E-31008 Pamplona, Spain
| | - Carlos Aydillo
- Facultad de Farmacia y Nutrición, Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea, E-31008 Pamplona, Spain; (D.P.); (S.E.); (E.M.); (C.A.); (C.S.)
- Instituto de Salud Tropical, Universidad de Navarra, ISTUN, Irunlarrea, E-31008 Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA) Irunlarrea, E-31008 Pamplona, Spain
| | - Carmen Sanmartín
- Facultad de Farmacia y Nutrición, Departamento de Tecnología y Química Farmacéuticas, Universidad de Navarra, Irunlarrea, E-31008 Pamplona, Spain; (D.P.); (S.E.); (E.M.); (C.A.); (C.S.)
- Instituto de Salud Tropical, Universidad de Navarra, ISTUN, Irunlarrea, E-31008 Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA) Irunlarrea, E-31008 Pamplona, Spain
| | - Manuel Sánchez-Moreno
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios de Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain; (D.M.-C.); (M.J.R.); (M.S.-M.)
| | - Clotilde Marín
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios de Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain; (D.M.-C.); (M.J.R.); (M.S.-M.)
- Correspondence: (R.M.-E.); (C.M.)
| |
Collapse
|
32
|
Mazzeti AL, Capelari-Oliveira P, Bahia MT, Mosqueira VCF. Review on Experimental Treatment Strategies Against Trypanosoma cruzi. J Exp Pharmacol 2021; 13:409-432. [PMID: 33833592 PMCID: PMC8020333 DOI: 10.2147/jep.s267378] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
Chagas disease is a neglected tropical disease caused by the protozoan Trypanosoma cruzi. Currently, only nitroheterocyclic nifurtimox (NFX) and benznidazole (BNZ) are available for the treatment of Chagas disease, with limitations such as variable efficacy, long treatment regimens and toxicity. Different strategies have been used to discover new active molecules for the treatment of Chagas disease. Target-based and phenotypic screening led to thousands of compounds with anti-T. cruzi activity, notably the nitroheterocyclic compounds, fexinidazole and its metabolites. In addition, drug repurposing, drug combinations, re-dosing regimens and the development of new formulations have been evaluated. The CYP51 antifungal azoles, as posaconazole, ravuconazole and its prodrug fosravuconazole presented promising results in experimental Chagas disease. Drug combinations of nitroheterocyclic and azoles were able to induce cure in murine infection. New treatment schemes using BNZ showed efficacy in the experimental chronic stage, including against dormant forms of T. cruzi. And finally, sesquiterpene lactone formulated in nanocarriers displayed outstanding efficacy against different strains of T. cruzi, susceptible or resistant to BNZ, the reference drug. These pre-clinical results are encouraging and provide interesting evidence to improve the treatment of patients with Chagas disease.
Collapse
Affiliation(s)
- Ana Lia Mazzeti
- Laboratório de Desenvolvimento Galênico e Nanotecnologia, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil.,Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, 21040-360, Brazil.,Laboratório de Doenças Parasitárias, Escola de Medicina & Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Patricia Capelari-Oliveira
- Laboratório de Desenvolvimento Galênico e Nanotecnologia, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Maria Terezinha Bahia
- Laboratório de Doenças Parasitárias, Escola de Medicina & Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Vanessa Carla Furtado Mosqueira
- Laboratório de Desenvolvimento Galênico e Nanotecnologia, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| |
Collapse
|
33
|
Zuma AA, de Souza W. Chagas Disease Chemotherapy: What Do We Know So Far? Curr Pharm Des 2021; 27:3963-3995. [PMID: 33593251 DOI: 10.2174/1381612827666210216152654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/13/2021] [Indexed: 11/22/2022]
Abstract
Chagas disease is a Neglected Tropical Disease (NTD), and although endemic in Latin America, affects around 6-7 million people infected worldwide. The treatment of Chagas disease is based on benznidazole and nifurtimox, which are the only available drugs. However, they are not effective during the chronic phase and cause several side effects. Furthermore, BZ promotes cure in 80% of the patients in the acute phase, but the cure rate drops to 20% in adults in the chronic phase of the disease. In this review, we present several studies published in the last six years, which describes the antiparasitic potential of distinct drugs, from the synthesis of new compounds aiming to target the parasite, as well as the repositioning and the combination of drugs. We highlight several compounds for having shown results that are equivalent or superior to BZ, which means that they should be further studied, either in vitro or in vivo. Furthermore, we stand out the differences in the effects of BZ on the same strain of T. cruzi, which might be related to methodological differences such as parasite and cell ratios, host cell type and the time of adding the drug. In addition, we discuss the wide variety of strains and also the cell types used as a host cell, which makes it difficult to compare the trypanocidal effect of the compounds.
Collapse
Affiliation(s)
- Aline Araujo Zuma
- Laboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho, 373, Centro de Ciências da Saúde, Cidade Universitária, Ilha do Fundão, 21491-590, Rio de Janeiro, RJ. Brazil
| | - Wanderley de Souza
- Laboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro. Av. Carlos Chagas Filho, 373, Centro de Ciências da Saúde, Cidade Universitária, Ilha do Fundão, 21491-590, Rio de Janeiro, RJ. Brazil
| |
Collapse
|
34
|
Pharmacokinetics of Benznidazole in Experimental Chronic Chagas Disease Using the Swiss Mouse-Berenice-78 Trypanosoma cruzi Strain Model. Antimicrob Agents Chemother 2021; 65:AAC.01383-20. [PMID: 33168611 DOI: 10.1128/aac.01383-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/04/2020] [Indexed: 01/01/2023] Open
Abstract
Chronic Chagas disease might have an impact on benznidazole pharmacokinetics with potential alterations in the therapeutic dosing regimen. This study aims to investigate the influence of chronic Trypanosoma cruzi infection on the pharmacokinetics and biodistribution of benznidazole in mice. Healthy (n = 40) and chronically T. cruzi (Berenice-78 strain)-infected (n = 40) Swiss female 10-month-old mice received a single oral dose of 100 mg/kg of body weight of benznidazole. Serial blood, heart, colon, and brain samples were collected up to 12 h after benznidazole administration. The serum and tissue samples were analyzed using a high-performance liquid chromatography instrument coupled to a diode array detector. Chronic infection by T. cruzi increased the values of the pharmacokinetic parameters absorption rate constant (Ka ) (3.92 versus 1.82 h-1), apparent volume of distribution (V/F) (0.089 versus 0.036 liters), and apparent clearance (CL/F) (0.030 versus 0.011 liters/h) and reduced the values of the time to the maximum concentration of drug in serum (T max) (0.67 versus 1.17 h) and absorption half-life (t 1/2 a ) (0.18 versus 0.38 h). Tissue exposure (area under the concentration-versus-time curve from 0 h to time t for tissue [AUC0- t ,tissue]) was longer and higher in the colon (8.15 versus 21.21 μg · h/g) and heart (5.72 versus 13.58 μg · h/g) of chronically infected mice. Chronic infection also increased the benznidazole tissue penetration ratios (AUC0- t ,tissue/AUC0- t ,serum ratios) of brain, colon, and heart by 1.6-, 3.25-, and 3-fold, respectively. The experimental chronic Chagas disease inflammation-mediated changes in the regulation of membrane transporters probably influence the benznidazole pharmacokinetics and the extent of benznidazole exposure in tissues. These results advise for potential alterations in benznidazole pharmacokinetics in chronic Chagas disease patients with possibilities of changes in the standard dosing regimen.
Collapse
|
35
|
Martín-Escolano R, Guardia JJ, Martín-Escolano J, Cirauqui N, Fernández A, Rosales MJ, Chahboun R, Sánchez-Moreno M, Alvarez-Manzaneda E, Marín C. In Vivo Biological Evaluation of a Synthetic Royleanone Derivative as a Promising Fast-Acting Trypanocidal Agent by Inducing Mitochondrial-Dependent Necrosis. JOURNAL OF NATURAL PRODUCTS 2020; 83:3571-3583. [PMID: 33253573 DOI: 10.1021/acs.jnatprod.0c00651] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The life-long and life-threatening Chagas disease is one of the most neglected tropical diseases caused by the protozoan parasite Trypanosoma cruzi. It is a major public health problem in Latin America, as six to seven million people are infected, being the principal cause of mortality in many endemic regions. Moreover, Chagas disease has become widespread due to migrant populations. Additionally, there are no vaccines nor effective treatments to fight the disease because of its long-term nature and complex pathology. Therefore, these facts emphasize how crucial the international effort for the development of new treatments against Chagas disease is. Here, we present the in vitro and in vivo trypanocidal activity of some oxygenated abietane diterpenoids and related compounds. The 1,4-benzoquinone 15, not yet reported, was identified as a fast-acting trypanocidal drug with efficacy against different strains in vitro and higher activity and lower toxicity than benznidazole in both phases of murine Chagas disease. The mode of action was also evaluated, suggesting that quinone 15 kills T. cruzi by inducing mitochondrion-dependent necrosis through a bioenergetics collapse caused by a mitochondrial membrane depolarization and iron-containing superoxide dismutase inhibition. Therefore, the abietane 1,4-benzoquinone 15 can be considered as a new candidate molecule for the development of an appropriate and commercially accessible anti-Chagas drug.
Collapse
Affiliation(s)
- Rubén Martín-Escolano
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | - Juan J Guardia
- Departamento de Química Orgánica, Facultad de Ciencias, Instituto de Biotecnología, Universidad de Granada, 18071 Granada, Spain
| | - Javier Martín-Escolano
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | - Nuria Cirauqui
- Molecular Microbiology and Structural Biochemistry, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, 69367 Lyon Cedex 07, France
| | - Antonio Fernández
- Departamento de Química Orgánica, Facultad de Ciencias, Instituto de Biotecnología, Universidad de Granada, 18071 Granada, Spain
| | - Maria J Rosales
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | - Rachid Chahboun
- Departamento de Química Orgánica, Facultad de Ciencias, Instituto de Biotecnología, Universidad de Granada, 18071 Granada, Spain
| | - Manuel Sánchez-Moreno
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | - Enrique Alvarez-Manzaneda
- Departamento de Química Orgánica, Facultad de Ciencias, Instituto de Biotecnología, Universidad de Granada, 18071 Granada, Spain
| | - Clotilde Marín
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| |
Collapse
|
36
|
Martín-Escolano J, Medina-Carmona E, Martín-Escolano R. Chagas Disease: Current View of an Ancient and Global Chemotherapy Challenge. ACS Infect Dis 2020; 6:2830-2843. [PMID: 33034192 DOI: 10.1021/acsinfecdis.0c00353] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Chagas disease is a neglected tropical disease and a global public health issue. In terms of treatment, no progress has been made since the 1960s, when benznidazole and nifurtimox, two obsolete drugs still prescribed, were used to treat this disease. Hence, currently, there are no effective treatments available to tackle Chagas disease. Over the past 20 years, there has been an increasing interest in the disease. However, parasite genetic diversity, drug resistance, tropism, and complex life cycle, along with the limited understanding of the disease and inadequate methodologies and strategies, have resulted in the absence of new insights in drugs development and disappointing outcomes in clinical trials so far. In summary, new drugs are urgently needed. This Review considers the relevant aspects related to the lack of drugs for Chagas disease, resumes the advances in tools for drug discovery, and discusses the main features to be taken into account to develop new effective drugs.
Collapse
Affiliation(s)
- Javier Martín-Escolano
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | | | - Rubén Martín-Escolano
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| |
Collapse
|
37
|
Thompson AM, O'Connor PD, Marshall AJ, Francisco AF, Kelly JM, Riley J, Read KD, Perez CJ, Cornwall S, Thompson RCA, Keenan M, White KL, Charman SA, Zulfiqar B, Sykes ML, Avery VM, Chatelain E, Denny WA. Re-evaluating pretomanid analogues for Chagas disease: Hit-to-lead studies reveal both in vitro and in vivo trypanocidal efficacy. Eur J Med Chem 2020; 207:112849. [PMID: 33007723 DOI: 10.1016/j.ejmech.2020.112849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/08/2020] [Accepted: 09/12/2020] [Indexed: 01/08/2023]
Abstract
Phenotypic screening of a 900 compound library of antitubercular nitroimidazole derivatives related to pretomanid against the protozoan parasite Trypanosoma cruzi (the causative agent for Chagas disease) identified several structurally diverse hits with an unknown mode of action. Following initial profiling, a first proof-of-concept in vivo study was undertaken, in which once daily oral dosing of a 7-substituted 2-nitroimidazooxazine analogue suppressed blood parasitemia to low or undetectable levels, although sterile cure was not achieved. Limited hit expansion studies alongside counter-screening of new compounds targeted at visceral leishmaniasis laid the foundation for a more in-depth assessment of the best leads, focusing on both drug-like attributes (solubility, metabolic stability and safety) and maximal killing of the parasite in a shorter timeframe. Comparative appraisal of one preferred lead (58) in a chronic infection mouse model, monitored by highly sensitive bioluminescence imaging, provided the first definitive evidence of (partial) curative efficacy with this promising nitroimidazooxazine class.
Collapse
Affiliation(s)
- Andrew M Thompson
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| | - Patrick D O'Connor
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Andrew J Marshall
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Amanda F Francisco
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, United Kingdom
| | - John M Kelly
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, United Kingdom
| | - Jennifer Riley
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, United Kingdom
| | - Kevin D Read
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, United Kingdom
| | - Catherine J Perez
- Department of Parasitology & Veterinary Sciences, Murdoch University, South Street, Murdoch, Western Australia, 6150, Australia
| | - Scott Cornwall
- Department of Parasitology & Veterinary Sciences, Murdoch University, South Street, Murdoch, Western Australia, 6150, Australia
| | - R C Andrew Thompson
- Department of Parasitology & Veterinary Sciences, Murdoch University, South Street, Murdoch, Western Australia, 6150, Australia
| | - Martine Keenan
- Epichem Pty Ltd, Suite 5, 3 Brodie-Hall Drive, Technology Park, Bentley, Western Australia, 6102, Australia
| | - Karen L White
- Centre for Drug Candidate Optimisation, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, Australia
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, Australia
| | - Bilal Zulfiqar
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland, 4111, Australia
| | - Melissa L Sykes
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland, 4111, Australia
| | - Vicky M Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland, 4111, Australia
| | - Eric Chatelain
- Drugs for Neglected Diseases initiative, 15 Chemin Louis Dunant, 1202, Geneva, Switzerland
| | - William A Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| |
Collapse
|
38
|
Scarim CB, Chin CM. Current Approaches to Drug Discovery for Chagas Disease: Methodological Advances. Comb Chem High Throughput Screen 2020; 22:509-520. [PMID: 31608837 DOI: 10.2174/1386207322666191010144111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/31/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND In recent years, there has been an improvement in the in vitro and in vivo methodology for the screening of anti-chagasic compounds. Millions of compounds can now have their activity evaluated (in large compound libraries) by means of high throughput in vitro screening assays. OBJECTIVE Current approaches to drug discovery for Chagas disease. METHOD This review article examines the contribution of these methodological advances in medicinal chemistry in the last four years, focusing on Trypanosoma cruzi infection, obtained from the PubMed, Web of Science, and Scopus databases. RESULTS Here, we have shown that the promise is increasing each year for more lead compounds for the development of a new drug against Chagas disease. CONCLUSION There is increased optimism among those working with the objective to find new drug candidates for optimal treatments against Chagas disease.
Collapse
Affiliation(s)
- Cauê B Scarim
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, Brazil.,Lapdesf - Laboratory of Research and Development of Drugs, Araraquara, São Paulo, Brazil
| | - Chung M Chin
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, Brazil.,Lapdesf - Laboratory of Research and Development of Drugs, Araraquara, São Paulo, Brazil
| |
Collapse
|
39
|
Cafferata ML, Toscani MA, Althabe F, Belizán JM, Bergel E, Berrueta M, Capparelli EV, Ciganda Á, Danesi E, Dumonteil E, Gibbons L, Gulayin PE, Herrera C, Momper JD, Rossi S, Shaffer JG, Schijman AG, Sosa-Estani S, Stella CB, Klein K, Buekens P. Short-course Benznidazole treatment to reduce Trypanosoma cruzi parasitic load in women of reproductive age (BETTY): a non-inferiority randomized controlled trial study protocol. Reprod Health 2020; 17:128. [PMID: 32831069 PMCID: PMC7446054 DOI: 10.1186/s12978-020-00972-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Retrospective observational studies suggest that transmission of Trypanosoma cruzi does not occur in treated women when pregnant later in life. The level of parasitemia is a known risk factor for congenital transmission. Benznidazole (BZN) is the drug of choice for preconceptional treatment to reduce parasitic load. The fear of treatment-related side effects limits the implementation of the Argentine guideline recommending BZN 60d/300 mg (or equivalent) treatment of T. cruzi seropositive women during the postpartum period to prevent transmission in a future pregnancy. A short and low dose BZN treatment might reduce major side effects and increase compliance, but its efficacy to reduce T. cruzi parasitic load compared to the standard 60d/300 mg course is not yet established. Clinical trials testing alternative BZN courses among women of reproductive age are urgently needed. METHODS AND DESIGN We are proposing to perform a double-blinded, non-inferiority randomized controlled trial comparing a short low dose 30-day treatment with BZN 150 mg/day (30d/150 mg) vs. BZN 60d/300 mg. We will recruit not previously treated T. cruzi seropositive women with a live birth during the postpartum period in Argentina, randomize them at 6 months postpartum, and follow them up with the following specific aims: Specific aim 1: to measure the effect of BZN 30d/150 mg compared to 60d/300 mg preconceptional treatment on parasitic load measured by the frequency of positive Polymerase Chain Reaction (PCR) (primary outcome) and by real-time quantitative PCR (qPCR), immediately and 10 months after treatment. Specific aim 2: to measure the frequency of serious adverse events and/or any adverse event leading to treatment interruption. TRIAL REGISTRATION ClinicalTrials.gov . Identifier: NCT03672487 . Registered 14 September 2018.
Collapse
Affiliation(s)
- María L Cafferata
- Instituto de Efectividad Clinica y Sanitaria (IECS), Buenos Aires, Argentina.
- Unidad de Investigación Clínica y Epidemiológica Montevideo (UNICEM), Montevideo, Uruguay.
| | - María A Toscani
- Instituto de Efectividad Clinica y Sanitaria (IECS), Buenos Aires, Argentina
| | - Fernando Althabe
- Maternal and Perinatal Health. UNDP-UNFPA-UNICEF-WHO-World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP), Department of Sexual and Reproductive Health and Research (SRH), World Health Organization, Geneva, Switzerland
| | - Jose M Belizán
- Instituto de Efectividad Clinica y Sanitaria (IECS), Buenos Aires, Argentina
| | - Eduardo Bergel
- Instituto de Efectividad Clinica y Sanitaria (IECS), Buenos Aires, Argentina
| | - Mabel Berrueta
- Instituto de Efectividad Clinica y Sanitaria (IECS), Buenos Aires, Argentina
| | - Edmund V Capparelli
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego (UCSD), San Diego, USA
- Schools of Medicine, University of California, San Diego (UCSD), San Diego, USA
| | - Álvaro Ciganda
- Instituto de Efectividad Clinica y Sanitaria (IECS), Buenos Aires, Argentina
- Unidad de Investigación Clínica y Epidemiológica Montevideo (UNICEM), Montevideo, Uruguay
| | - Emmaria Danesi
- Centro Nacional de Diagnóstico e Investigación en Endemoepidemias (CeNDIE) ANLIS Dr. C. G. Malbrán, Buenos Aires, Argentina
| | - Eric Dumonteil
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| | - Luz Gibbons
- Instituto de Efectividad Clinica y Sanitaria (IECS), Buenos Aires, Argentina
| | - Pablo E Gulayin
- Instituto de Efectividad Clinica y Sanitaria (IECS), Buenos Aires, Argentina
| | - Claudia Herrera
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego (UCSD), San Diego, USA
| | - Steven Rossi
- Schools of Medicine, University of California, San Diego (UCSD), San Diego, USA
| | - Jeffrey G Shaffer
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| | - Alejandro G Schijman
- Laboratorio de Biología Molecular de la Enfermedad de Chagas (LaBMECh), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor Torres" (INGEBI), Buenos Aires, Argentina
| | - Sergio Sosa-Estani
- Drugs for Neglected Diseases initiative - Latin America (DNDi), Rio de Janeiro, Brazil
- Centro de Investigaciones en Epidemiología y Salud Pública (CIESP-IECS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Candela B Stella
- Instituto de Efectividad Clinica y Sanitaria (IECS), Buenos Aires, Argentina
| | - Karen Klein
- Instituto de Efectividad Clinica y Sanitaria (IECS), Buenos Aires, Argentina
| | - Pierre Buekens
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| |
Collapse
|
40
|
Chatelain E, Scandale I. Animal models of Chagas disease and their translational value to drug development. Expert Opin Drug Discov 2020; 15:1381-1402. [PMID: 32812830 DOI: 10.1080/17460441.2020.1806233] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION American trypanosomiasis, better known as Chagas disease, is a global public health issue. Current treatments targeting the causative parasite, Trypanosoma cruzi, are limited to two old nitroheterocyclic compounds; new, safer drugs are needed. New tools to identify compounds suitable for parasitological cure in humans have emerged through efforts in drug discovery. AREAS COVERED Animal disease models are an integral part of the drug discovery process. There are numerous experimental models of Chagas disease described and in use; rather than going through each of these and their specific features, the authors focus on developments in recent years, in particular the imaging technologies that have dramatically changed the Chagas R&D landscape, and provide a critical view on their value and limitations for moving compounds forward into further development. EXPERT OPINION The application of new technological advances to the field of drug development for Chagas disease has led to the implementation of new and robust/standardized in vivo models that contributed to a better understanding of host/parasite interactions. These new models should also build confidence in their translational value for moving compounds forward into clinical development.
Collapse
Affiliation(s)
- Eric Chatelain
- R&D Department, Drugs for Neglected Diseases Initiative (DNDi) , Geneva, Switzerland
| | - Ivan Scandale
- R&D Department, Drugs for Neglected Diseases Initiative (DNDi) , Geneva, Switzerland
| |
Collapse
|
41
|
Abstract
Infections with Trypanosoma cruzi are usually lifelong despite generating a strong adaptive immune response. Identifying the sites of parasite persistence is therefore crucial to understanding how T. cruzi avoids immune-mediated destruction. However, this is a major technical challenge, because the parasite burden during chronic infections is extremely low. Here, we describe an integrated approach involving comprehensive tissue processing, ex vivo imaging, and confocal microscopy, which allowed us to visualize infected host cells in murine tissue with exquisite sensitivity. Using bioluminescence-guided tissue sampling, with a detection level of <20 parasites, we showed that in the colon, smooth muscle myocytes in the circular muscle layer are the most common infected host cell type. Typically, during chronic infections, the entire colon of a mouse contains only a few hundred parasites, often concentrated in a small number of cells each containing >200 parasites, which we term mega-nests. In contrast, during the acute stage, when the total parasite burden is considerably higher and many cells are infected, nests containing >50 parasites are rarely found. In C3H/HeN mice, but not BALB/c mice, we identified skeletal muscle as a major site of persistence during the chronic stage, with most parasites being found in large mega-nests within the muscle fibers. Finally, we report that parasites are also frequently found in the skin during chronic murine infections, often in multiple infection foci. In addition to being a site of parasite persistence, this anatomical reservoir could play an important role in insect-mediated transmission and have implications for drug development.IMPORTANCE Trypanosoma cruzi causes Chagas disease, the most important parasitic infection in Latin America. Major pathologies include severe damage to the heart and digestive tract, although symptoms do not usually appear until decades after infection. Research has been hampered by the complex nature of the disease and technical difficulties in locating the extremely low number of parasites. Here, using highly sensitive imaging technology, we reveal the sites of parasite persistence during chronic-stage infections of experimental mice at single-cell resolution. We show that parasites are frequently located in smooth muscle cells in the circular muscle layer of the colon and that skeletal muscle cells and the skin can also be important reservoirs. This information provides a framework for investigating how the parasite is able to survive as a lifelong infection, despite a vigorous immune response. It also informs drug development strategies by identifying tissue sites that must be accessed to achieve a curative outcome.
Collapse
|
42
|
Francisco AF, Jayawardhana S, Olmo F, Lewis MD, Wilkinson SR, Taylor MC, Kelly JM. Challenges in Chagas Disease Drug Development. Molecules 2020; 25:E2799. [PMID: 32560454 PMCID: PMC7355550 DOI: 10.3390/molecules25122799] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 02/08/2023] Open
Abstract
The protozoan parasite Trypanosoma cruzi causes Chagas disease, an important public health problem throughout Latin America. Current therapeutic options are characterised by limited efficacy, long treatment regimens and frequent toxic side-effects. Advances in this area have been compromised by gaps in our knowledge of disease pathogenesis, parasite biology and drug activity. Nevertheless, several factors have come together to create a more optimistic scenario. Drug-based research has become more systematic, with increased collaborations between the academic and commercial sectors, often within the framework of not-for-profit consortia. High-throughput screening of compound libraries is being widely applied, and new technical advances are helping to streamline the drug development pipeline. In addition, drug repurposing and optimisation of current treatment regimens, informed by laboratory research, are providing a basis for new clinical trials. Here, we will provide an overview of the current status of Chagas disease drug development, highlight those areas where progress can be expected, and describe how fundamental research is helping to underpin the process.
Collapse
Affiliation(s)
- Amanda F. Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Shane R. Wilkinson
- School of Biological and Chemical Sciences, Queen Mary University of London Mile End Road, London E1 4NS, UK;
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| |
Collapse
|
43
|
Pereira CA, Sayé M, Reigada C, Silber AM, Labadie GR, Miranda MR, Valera-Vera E. Computational approaches for drug discovery against trypanosomatid-caused diseases. Parasitology 2020; 147:611-633. [PMID: 32046803 PMCID: PMC10317681 DOI: 10.1017/s0031182020000207] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
During three decades, only about 20 new drugs have been developed for malaria, tuberculosis and all neglected tropical diseases (NTDs). This critical situation was reached because NTDs represent only 10% of health research investments; however, they comprise about 90% of the global disease burden. Computational simulations applied in virtual screening (VS) strategies are very efficient tools to identify pharmacologically active compounds or new indications for drugs already administered for other diseases. One of the advantages of this approach is the low time-consuming and low-budget first stage, which filters for testing experimentally a group of candidate compounds with high chances of binding to the target and present trypanocidal activity. In this work, we review the most common VS strategies that have been used for the identification of new drugs with special emphasis on those applied to trypanosomiasis and leishmaniasis. Computational simulations based on the selected protein targets or their ligands are explained, including the method selection criteria, examples of successful VS campaigns applied to NTDs, a list of validated molecular targets for drug development and repositioned drugs for trypanosomatid-caused diseases. Thereby, here we present the state-of-the-art of VS and drug repurposing to conclude pointing out the future perspectives in the field.
Collapse
Affiliation(s)
- Claudio A. Pereira
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Melisa Sayé
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Chantal Reigada
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Ariel M. Silber
- Laboratory of Biochemistry of Tryps – LaBTryps, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Guillermo R. Labadie
- Instituto de Química Rosario (IQUIR-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
- Departamento de Química Orgánica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Mariana R. Miranda
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Edward Valera-Vera
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| |
Collapse
|
44
|
Drug-cured experimental Trypanosoma cruzi infections confer long-lasting and cross-strain protection. PLoS Negl Trop Dis 2020; 14:e0007717. [PMID: 32302312 PMCID: PMC7190179 DOI: 10.1371/journal.pntd.0007717] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 04/29/2020] [Accepted: 02/11/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The long term and complex nature of Chagas disease in humans has restricted studies on vaccine feasibility. Animal models also have limitations due to technical difficulties in monitoring the extremely low parasite burden that is characteristic of chronic stage infections. Advances in imaging technology offer alternative approaches that circumvent these problems. Here, we describe the use of highly sensitive whole body in vivo imaging to assess the efficacy of recombinant viral vector vaccines and benznidazole-cured infections to protect mice from challenge with Trypanosoma cruzi. METHODOLOGY/PRINCIPAL FINDINGS Mice were infected with T. cruzi strains modified to express a red-shifted luciferase reporter. Using bioluminescence imaging, we assessed the degree of immunity to re-infection conferred after benznidazole-cure. Those infected for 14 days or more, prior to the onset of benznidazole treatment, were highly protected from challenge with both homologous and heterologous strains. There was a >99% reduction in parasite burden, with parasites frequently undetectable after homologous challenge. This level of protection was considerably greater than that achieved with recombinant vaccines. It was also independent of the route of infection or size of the challenge inoculum, and was long-lasting, with no significant diminution in immunity after almost a year. When the primary infection was benznidazole-treated after 4 days (before completion of the first cycle of intracellular infection), the degree of protection was much reduced, an outcome associated with a minimal T. cruzi-specific IFN-γ+ T cell response. CONCLUSIONS/SIGNIFICANCE Our findings suggest that a protective Chagas disease vaccine must have the ability to eliminate parasites before they reach organs/tissues, such as the GI tract, where once established, they become largely refractory to the induced immune response.
Collapse
|
45
|
High Throughput Approaches to Unravel the Mechanism of Action of a New Vanadium-Based Compound against Trypanosoma cruzi. Bioinorg Chem Appl 2020; 2020:1634270. [PMID: 32351549 PMCID: PMC7171612 DOI: 10.1155/2020/1634270] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/03/2020] [Indexed: 12/17/2022] Open
Abstract
Treatment for Chagas disease, a parasitosis caused by Trypanosoma cruzi, has always been based on two drugs, nifurtimox and benznidazole, despite the toxic side effects described after prolonged prescription. In this work, we study a new prospective antitrypanosomal drug based on vanadium, here named VIVO(5Brsal)(aminophen). We found a good IC50 value, (3.76 ± 0.08) μM, on CL Brener epimastigotes. The analysis of cell death mechanism allowed us to rule out the implication of a mechanism based on early apoptosis or necrosis. Recovery assays revealed a trypanostatic effect, accompanied by cell shape and motility alterations. An uptake mostly associated with the insoluble fraction of the parasites was deduced through vanadium determinations. Concordantly, no drastic changes of the parasite transcriptome were detected after 6 h of treatment. Instead, proteomic analysis uncovered the modulation of proteins involved in different processes such as energy and redox metabolism, transport systems, detoxifying pathways, ribosomal protein synthesis, and proteasome protein degradation. Overall, the results here presented lead us to propose that VIVO(5Brsal)(aminophen) exerts a trypanostatic effect on T. cruzi affecting parasite insoluble proteins.
Collapse
|
46
|
Ribeiro V, Dias N, Paiva T, Hagström-Bex L, Nitz N, Pratesi R, Hecht M. Current trends in the pharmacological management of Chagas disease. Int J Parasitol Drugs Drug Resist 2020; 12:7-17. [PMID: 31862616 PMCID: PMC6928327 DOI: 10.1016/j.ijpddr.2019.11.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/06/2019] [Accepted: 11/28/2019] [Indexed: 12/12/2022]
Abstract
Chagas disease (CD) is a tropical neglected illness, affecting mainly populations of low socioeconomic status in Latin America. An estimated 6 to 8 million people worldwide are infected with Trypanosoma cruzi, the etiological agent of CD. Despite being one of the main global health problems, this disease continues without effective treatment during the chronic phase of the infection. The limitation of therapeutic strategies has been one of the biggest challenges on the fight against CD. Nifurtimox and benznidazole, developed in the 1970s, are still the only commercial options with established efficacy on CD. However, the efficacy of these drugs have a proven efficacy only during early infection and the benefits in the chronic phase are questionable. Consequently, there is a growing need for new pharmacological alternatives, either by optimization of existing drugs or by the formulation of new compounds. In the present study, a literature review of the currently adopted therapy, its concomitant combination with other drugs, and potential future treatments for CD was performed, considering articles published from 2012. The revised articles were selected according to the protocol of treatment: evaluation of drug association, drug repositioning and research of new drugs. As a result of the present revision, it was possible to conclude that the use of benznidazole in combination with other compounds showed better results when compared with its use as a single therapy. The search of new drugs has been the strategy most used in pursuing more effective forms of treatment for CD. However, studies have still focused on basic research, that is, they are still in a pre-clinical stage, using methodologies based on in vitro or in animal studies.
Collapse
Affiliation(s)
- Vanessa Ribeiro
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasilia, Federal District, Brazil.
| | - Nayra Dias
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasilia, Federal District, Brazil.
| | - Taís Paiva
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasilia, Federal District, Brazil.
| | - Luciana Hagström-Bex
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasilia, Federal District, Brazil.
| | - Nadjar Nitz
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasilia, Federal District, Brazil.
| | - Riccardo Pratesi
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasilia, Federal District, Brazil.
| | - Mariana Hecht
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasilia, Federal District, Brazil.
| |
Collapse
|
47
|
Taylor MC, Ward A, Olmo F, Jayawardhana S, Francisco AF, Lewis MD, Kelly JM. Intracellular DNA replication and differentiation of Trypanosoma cruzi is asynchronous within individual host cells in vivo at all stages of infection. PLoS Negl Trop Dis 2020; 14:e0008007. [PMID: 32196491 PMCID: PMC7112235 DOI: 10.1371/journal.pntd.0008007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/01/2020] [Accepted: 02/29/2020] [Indexed: 12/21/2022] Open
Abstract
Investigations into intracellular replication and differentiation of Trypanosoma cruzi within the mammalian host have been restricted by limitations in our ability to detect parasitized cells throughout the course of infection. We have overcome this problem by generating genetically modified parasites that express a bioluminescent/fluorescent fusion protein. By combining in vivo imaging and confocal microscopy, this has enabled us to routinely visualise murine infections at the level of individual host cells. These studies reveal that intracellular parasite replication is an asynchronous process, irrespective of tissue location or disease stage. Furthermore, using TUNEL assays and EdU labelling, we demonstrate that within individual infected cells, replication of both mitochondrial (kDNA) and nuclear genomes is not co-ordinated within the parasite population, and that replicating amastigotes and non-replicating trypomastigotes can co-exist in the same cell. Finally, we report the presence of distinct non-canonical morphological forms of T. cruzi in the mammalian host. These appear to represent transitional forms in the amastigote to trypomastigote differentiation process. Therefore, the intracellular life-cycle of T. cruzi in vivo is more complex than previously realised, with potential implications for our understanding of disease pathogenesis, immune evasion and drug development. Dissecting the mechanisms involved will be an important experimental challenge. Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, is becoming an emerging threat in non-endemic countries and establishing new foci in endemic countries. The treatment available has not changed significantly in over 40 years. Therefore, there is an urgent need for a greater understanding of parasite biology and disease pathogenesis to identify new therapeutic targets and to maximise the efficient use of existing drugs. We have used genetically modified strains of T. cruzi carrying a bioluminescence/fluorescence dual reporter fusion gene to monitor parasite replication in vivo during both acute and chronic infections in a mouse model. Utilising TUNEL assays for mitochondrial DNA replication and EdU incorporation for total DNA replication, we have found that parasite division within infected cells is asynchronous in all phases of infection. Differentiation also appears to be uncoordinated, with replicating amastigotes co-existing with non-dividing trypomastigotes in the same host cell.
Collapse
Affiliation(s)
- Martin C. Taylor
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- * E-mail:
| | - Alexander Ward
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Francisco Olmo
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Shiromani Jayawardhana
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Amanda F. Francisco
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
48
|
Molina I, Perin L, Aviles AS, de Abreu Vieira PM, da Silva Fonseca K, Cunha LM, Carneiro CM. The effect of benznidazole dose among the efficacy outcome in the murine animal model. A quantitative integration of the literature. Acta Trop 2020; 201:105218. [PMID: 31610148 DOI: 10.1016/j.actatropica.2019.105218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 09/06/2019] [Accepted: 10/10/2019] [Indexed: 12/17/2022]
Abstract
Despite more than 100 years since it was firstly described Chagas disease, only two drugs are available to treat Chagas disease: Nifurtimox launched by Bayer in 1965 and benznidazole launched by Roche in 1971. Drug discovery initiatives have been looking for new compounds as an alternative to these old drugs. Although new platforms have been used with the latest technologies, a critical step on that process still relies on the in vivo model. Unfortunately, to date, available animal models have limited predictive value and there is no standardization. With the aim to better understand the role of benznidazole, the current standard of care of Chagas disease, we performed this review. We intend to analyze the influence of the experimental design of the most used animal model, the murine model, in the assessment of the efficacy endpoint.
Collapse
Affiliation(s)
- Israel Molina
- Tropical Medicine Unit, Infectious Disease Department. PROSICS (International Health Program of the Catalan Health Institute), Hospital Universitario Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain; Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil.
| | - Luisa Perin
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Augusto Sao Aviles
- Tropical Medicine Unit, Infectious Disease Department. PROSICS (International Health Program of the Catalan Health Institute), Hospital Universitario Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Paula Melo de Abreu Vieira
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil; Laboratório de Morfopatologia, Departamento de Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Katia da Silva Fonseca
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Lucas Maciel Cunha
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Claudia M Carneiro
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| |
Collapse
|
49
|
Nitroheterocyclic derivatives: privileged scaffold for drug development against Chagas disease. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02453-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
50
|
Lin C, Hulpia F, da Silva CF, Batista DDGJ, Van Hecke K, Maes L, Caljon G, Soeiro MDNC, Van Calenbergh S. Discovery of Pyrrolo[2,3-b]pyridine (1,7-Dideazapurine) Nucleoside Analogues as Anti-Trypanosoma cruzi Agents. J Med Chem 2019; 62:8847-8865. [DOI: 10.1021/acs.jmedchem.9b01275] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Cai Lin
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000 Gent, Belgium
| | - Fabian Hulpia
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000 Gent, Belgium
| | - Cristiane França da Silva
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz (FIOCRUZ), Fundação Oswaldo Cruz, Rio de Janeiro, Avenida Brasil 4365, Manguinhos, 21040-360 Rio de Janeiro, Brazil
| | - Denise da Gama Jaen Batista
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz (FIOCRUZ), Fundação Oswaldo Cruz, Rio de Janeiro, Avenida Brasil 4365, Manguinhos, 21040-360 Rio de Janeiro, Brazil
| | - Kristof Van Hecke
- XStruct, Department of Chemistry, Ghent University, Krijgslaan 281 S3, B-9000 Gent, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610 Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610 Wilrijk, Belgium
| | - Maria de Nazaré C. Soeiro
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz (FIOCRUZ), Fundação Oswaldo Cruz, Rio de Janeiro, Avenida Brasil 4365, Manguinhos, 21040-360 Rio de Janeiro, Brazil
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, B-9000 Gent, Belgium
| |
Collapse
|