51
|
de Albuquerque S, Cianni L, de Vita D, Duque C, Gomes ASM, Gomes P, Laughton C, Leitão A, Montanari CA, Montanari R, Ribeiro JFR, da Silva JS, Teixeira C. Molecular design aided by random forests and synthesis of potent trypanocidal agents as cruzain inhibitors for Chagas disease treatment. Chem Biol Drug Des 2020; 96:948-960. [PMID: 33058457 DOI: 10.1111/cbdd.13663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/13/2019] [Accepted: 12/23/2019] [Indexed: 11/30/2022]
Abstract
Cruzain is an established target for the identification of novel trypanocidal agents, but how good are in vitro/in vivo correlations? This work describes the development of a random forests model for the prediction of the bioavailability of cruzain inhibitors that are Trypanosoma cruzi killers. Some common properties that characterize drug-likeness are poorly represented in many established cruzain inhibitors. This correlates with the evidence that many high-affinity cruzain inhibitors are not trypanocidal agents against T. cruzi. On the other hand, T. cruzi killers that present typical drug-like characteristics are likely to show better trypanocidal action than those without such features. The random forests model was not outperformed by other machine learning methods (such as artificial neural networks and support vector machines), and it was validated with the synthesis of two new trypanocidal agents. Specifically, we report a new lead compound, Neq0565, which was tested on T. cruzi Tulahuen (β-galactosidase) with a pEC50 of 4.9. It is inactive in the host cell line showing a selectivity index (SI = EC50 cyto /EC50 T. cruzi ) higher than 50.
Collapse
Affiliation(s)
- Sérgio de Albuquerque
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Lorenzo Cianni
- Grupo de Química Medicinal, Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos/SP, Brazil
| | - Daniela de Vita
- Grupo de Química Medicinal, Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos/SP, Brazil
| | - Carla Duque
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Ana S M Gomes
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Paula Gomes
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| | - Charles Laughton
- School of Pharmacy and Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Andrei Leitão
- Grupo de Química Medicinal, Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos/SP, Brazil
| | - Carlos A Montanari
- Grupo de Química Medicinal, Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos/SP, Brazil
| | - Raphael Montanari
- Centro de Robótica de São Carlos, EESC-ICMC, Universidade de São Paulo, São Paulo, Brazil
| | - Jean F R Ribeiro
- Grupo de Química Medicinal, Instituto de Química de São Carlos, Universidade de São Paulo, São Carlos/SP, Brazil
| | - João Santana da Silva
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Cátia Teixeira
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
| |
Collapse
|
52
|
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
|
53
|
Fernandes FS, Santos H, Lima SR, Conti C, Rodrigues MT, Zeoly LA, Ferreira LLG, Krogh R, Andricopulo AD, Coelho F. Discovery of highly potent and selective antiparasitic new oxadiazole and hydroxy-oxindole small molecule hybrids. Eur J Med Chem 2020; 201:112418. [PMID: 32590115 DOI: 10.1016/j.ejmech.2020.112418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 11/26/2022]
Abstract
A series of highly active hybrids were discovered as novel antiparasitic agents. Two heterocyclic scaffolds (1,2,4-oxadiazole and 3-hydroxy-2-oxindole) were linked, and the resulting compounds showed in vitro activities against intracellular amastigotes of two protozoan parasites, Trypanosoma cruzi and Leishmania infantum. Their cytotoxicity was assessed using HFF-1 fibroblasts and HepG2 hepatocytes. Compounds 5b, 5d, 8h and 8o showed selectivity against L. infantum (IC50 values of 3.89, 2.38, 2.50 and 2.85 μM, respectively). Compounds 4c, 4q, 8a and 8k were the most potent against T. cruzi, exhibiting IC50 values of 6.20, 2.20, 2.30 and 2.20 μM, respectively. Additionally, the most potent anti-T. cruzi compounds showed in vitro efficacies comparable or superior to that of benznidazole. These easy-to-synthesize molecules represent novel chemotypes for the design of potent and selective lead compounds for Chagas disease and leishmaniasis drug discovery.
Collapse
Affiliation(s)
- Fábio S Fernandes
- Laboratory of Synthesis of Natural Products and Drugs, Institute of Chemistry, University of Campinas, PO Box 6154, 13083-970, Campinas, SP, Brazil
| | - Hugo Santos
- Laboratory of Synthesis of Natural Products and Drugs, Institute of Chemistry, University of Campinas, PO Box 6154, 13083-970, Campinas, SP, Brazil
| | - Samia R Lima
- Laboratory of Synthesis of Natural Products and Drugs, Institute of Chemistry, University of Campinas, PO Box 6154, 13083-970, Campinas, SP, Brazil
| | - Caroline Conti
- Laboratory of Synthesis of Natural Products and Drugs, Institute of Chemistry, University of Campinas, PO Box 6154, 13083-970, Campinas, SP, Brazil
| | - Manoel T Rodrigues
- Laboratory of Synthesis of Natural Products and Drugs, Institute of Chemistry, University of Campinas, PO Box 6154, 13083-970, Campinas, SP, Brazil
| | - Lucas A Zeoly
- Laboratory of Synthesis of Natural Products and Drugs, Institute of Chemistry, University of Campinas, PO Box 6154, 13083-970, Campinas, SP, Brazil
| | - Leonardo L G Ferreira
- Laboratory of Medicinal and Computational Chemistry, Institute of Physics of Sao Carlos, University of Sao Paulo, Avenida Joao Dagnone, 1100, 13563-120, Sao Carlos, SP, Brazil
| | - Renata Krogh
- Laboratory of Medicinal and Computational Chemistry, Institute of Physics of Sao Carlos, University of Sao Paulo, Avenida Joao Dagnone, 1100, 13563-120, Sao Carlos, SP, Brazil
| | - Adriano D Andricopulo
- Laboratory of Medicinal and Computational Chemistry, Institute of Physics of Sao Carlos, University of Sao Paulo, Avenida Joao Dagnone, 1100, 13563-120, Sao Carlos, SP, Brazil.
| | - Fernando Coelho
- Laboratory of Synthesis of Natural Products and Drugs, Institute of Chemistry, University of Campinas, PO Box 6154, 13083-970, Campinas, SP, Brazil.
| |
Collapse
|
54
|
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: 27] [Impact Index Per Article: 5.4] [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
|
55
|
Fesser AF, Braissant O, Olmo F, Kelly JM, Mäser P, Kaiser M. Non-invasive monitoring of drug action: A new live in vitro assay design for Chagas' disease drug discovery. PLoS Negl Trop Dis 2020; 14:e0008487. [PMID: 32716934 PMCID: PMC7419005 DOI: 10.1371/journal.pntd.0008487] [Citation(s) in RCA: 5] [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: 01/15/2020] [Revised: 08/11/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023] Open
Abstract
New assay designs are needed to improve the predictive value of the Trypanosoma cruzi in vitro tests used as part of the Chagas' disease drug development pipeline. Here, we employed a green fluorescent protein (eGFP)-expressing parasite line and live high-content imaging to monitor the growth of T. cruzi amastigotes in mouse embryonic fibroblasts. A novel assay design allowed us to follow parasite numbers over 6 days, in four-hour intervals, while occupying the microscope for only 24 hours per biological replicate. Dose-response curves were calculated for each time point after addition of test compounds, revealing how EC50 values first decreased over the time of drug exposure, and then leveled off. However, we observed that parasite numbers could vary, even in the untreated controls, and at different sites in the same well, which caused variability in the EC50 values. To overcome this, we established that fold change in parasite number per hour is a more robust and informative measure of drug activity. This was calculated based on an exponential growth model for every biological sample. The net fold change per hour is the result of parasite replication, differentiation, and death. The calculation of this fold change enabled us to determine the tipping point of drug action, i.e. the time point when the death rate of the parasites exceeded the growth rate and the fold change dropped below 1, depending on the drug concentration and exposure time. This revealed specific pharmacodynamic profiles of the benchmark drugs benznidazole and posaconazole. Chagas' disease, caused by Trypanosoma cruzi, is a chronic debilitating infection occurring mostly in Latin America. There is an urgent need for new, well tolerated drugs. However, the latest therapeutic candidates have yielded disappointing outcomes in clinical trials, despite promising preclinical results. This demands new and more predictive in vitro assays. To address this, we have developed an assay design that enables the growth of T. cruzi intracellular forms to be monitored in real time, under drug pressure, for 6 days post-infection. This allowed us to establish the tipping point of drug action, when the death rate of the parasites exceeded the growth rate. The resulting pharmacodynamics profiles can provide robust and informative details on anti-chagasic candidates, as demonstrated for the benchmark drugs benznidazole and posaconazole.
Collapse
Affiliation(s)
- Anna F. Fesser
- Medical Parasitology and Infection Biology, Swiss Tropical & Public Health Institute, Switzerland
- University of Basel, Basel, Switzerland
| | - Olivier Braissant
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Francisco Olmo
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Pascal Mäser
- Medical Parasitology and Infection Biology, Swiss Tropical & Public Health Institute, Switzerland
- University of Basel, Basel, Switzerland
- * E-mail:
| | - Marcel Kaiser
- Medical Parasitology and Infection Biology, Swiss Tropical & Public Health Institute, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
56
|
Amlodipine Increases the Therapeutic Potential of Ravuconazole upon Trypanosoma cruzi Infection. Antimicrob Agents Chemother 2020; 64:AAC.02497-19. [PMID: 32423960 DOI: 10.1128/aac.02497-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/05/2020] [Indexed: 12/14/2022] Open
Abstract
Mining existing agents that enhance the therapeutic potential of ergosterol biosynthesis inhibitors (EBI) is a promising approach to improve Chagas disease chemotherapy. In this study, we evaluated the effect of ravuconazole, an EBI, combined with amlodipine, a calcium channel blocker, upon Trypanosoma cruzi experimental infection. In vitro assays confirmed the trypanocidal activity of both compounds in monotherapy and demonstrated an additive effect (sum of the fractional inhibitory concentration [ΣFIC] > 0.5) of the combined treatment without additional toxicity to host cells. In vivo experiments, using a murine model of the T. cruzi Y strain in a short-term protocol, demonstrated that amlodipine, although lacking trypanocidal activity, dramatically increased the antiparasitic activity of underdosing ravuconazole regimens. Additional analysis using long-term treatment (20 days) showed that parasitemia relapse until 60 days after treatment was significatively lower in mice treated with the combination (4 out of 14 mice) than ravuconazole monotherapy (10 out of 14 mice), even in the presence of immunosuppressant pressure. Furthermore, the combined therapy was well tolerated and protected the mice from mortality. The treatments also impacted on the cellular and humoral immune response of infected animals, inducing a reduction of serum cytokine levels in all ravuconazole-treated mice. Our findings demonstrate that amlodipine is efficacious in enhancing the antiparasitic activity of ravuconazole in an experimental model of T. cruzi infection and indicates a potential strategy to be explored in Chagas disease treatment.
Collapse
|
57
|
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: 32] [Impact Index Per Article: 6.4] [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
|
58
|
Franco CH, Warhurst DC, Bhattacharyya T, Au HYA, Le H, Giardini MA, Pascoalino BS, Torrecilhas AC, Romera LMD, Madeira RP, Schenkman S, Freitas-Junior LH, Chatelain E, Miles MA, Moraes CB. Novel structural CYP51 mutation in Trypanosoma cruzi associated with multidrug resistance to CYP51 inhibitors and reduced infectivity. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2020; 13:107-120. [PMID: 32688218 PMCID: PMC7369355 DOI: 10.1016/j.ijpddr.2020.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 12/31/2022]
Abstract
Ergosterol biosynthesis inhibitors, such as posaconazole and ravuconazole, have been proposed as drug candidates for Chagas disease, a neglected infectious tropical disease caused by the protozoan parasite Trypanosoma cruzi. To understand better the mechanism of action and resistance to these inhibitors, a clone of the T. cruzi Y strain was cultured under intermittent and increasing concentrations of ravuconazole until phenotypic stability was achieved. The ravuconazole-selected clone exhibited loss in fitness in vitro when compared to the wild-type parental clone, as observed in reduced invasion capacity and slowed population growth in both mammalian and insect stages of the parasite. In drug activity assays, the resistant clone was above 300-fold more tolerant to ravuconazole than the sensitive parental clone, when the half-maximum effective concentration (EC50) was considered. The resistant clones also showed reduced virulence in vivo, when compared to parental sensitive clones. Cross-resistance to posaconazole and other CYP51 inhibitors, but not to other antichagasic drugs that act independently of CYP51, such as benznidazole and nifurtimox, was also observed. A novel amino acid residue change, T297M, was found in the TcCYP51 gene in the resistant but not in the sensitive clones. The structural effects of the T297M, and of the previously described P355S residue changes, were modelled to understand their impact on interaction with CYP51 inhibitors. A ravuconazole-resistant T. cruzi clone presented reduced in vitro and in vivo fitness. The ravuconazole-resistant clone presented cross-resistance to other CYP51 inhibitors. There was no cross-resistance to benznidazole and nifurtimox. Resistance is associated with a novel structural mutation in the TcCYP51 protein.
Collapse
Affiliation(s)
- Caio H Franco
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP, Brazil; Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - David C Warhurst
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Tapan Bhattacharyya
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Ho Y A Au
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Hai Le
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Miriam A Giardini
- Institut Pasteur Korea, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Bruno S Pascoalino
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP, Brazil
| | - Ana Claudia Torrecilhas
- Department of Pharmaceutical Sciences, Federal University of São Paulo (UNIFESP), Diadema, SP, Brazil
| | - Lavinia M D Romera
- Department of Pharmaceutical Sciences, Federal University of São Paulo (UNIFESP), Diadema, SP, Brazil
| | - Rafael Pedro Madeira
- Department of Pharmaceutical Sciences, Federal University of São Paulo (UNIFESP), Diadema, SP, Brazil
| | - Sergio Schenkman
- Department of Microbiology, Immunology and Parasitology, UNIFESP, São Paulo, SP, Brazil
| | - Lucio H Freitas-Junior
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Institut Pasteur Korea, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Eric Chatelain
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | - Michael A Miles
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Carolina B Moraes
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP, Brazil; Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Institut Pasteur Korea, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea; Department of Pharmaceutical Sciences, Federal University of São Paulo (UNIFESP), Diadema, SP, Brazil.
| |
Collapse
|
59
|
Molina-Morant D, Fernández ML, Bosch-Nicolau P, Sulleiro E, Bangher M, Salvador F, Sanchez-Montalva A, Ribeiro ALP, de Paula AMB, Eloi S, Correa-Oliveira R, Villar JC, Sosa-Estani S, Molina I. Efficacy and safety assessment of different dosage of benznidazol for the treatment of Chagas disease in chronic phase in adults (MULTIBENZ study): study protocol for a multicenter randomized Phase II non-inferiority clinical trial. Trials 2020; 21:328. [PMID: 32293523 PMCID: PMC7158046 DOI: 10.1186/s13063-020-4226-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/03/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Chagas disease (CD) continues to be a neglected infectious disease with one of the largest burdens globally. Despite the modest cure rates in adult chronic patients and its safety profile, benznidazole (BNZ) is still the drug of choice. Its current recommended dose is based on nonrandomized studies, and efficacy and safety of the optimal dose of BNZ have been scarcely analyzed in clinical trials. METHODS/DESIGN MULTIBENZ is a phase II, randomized, noninferiority, double-blind, multicenter international clinical trial. A total of 240 patients with Trypanosoma CD in the chronic phase will be recruited in four different countries (Argentina, Brazil, Colombia, and Spain). Patients will be randomized to receive BNZ 150 mg/day for 60 days, 400 mg/day for 15 days, or 300 mg/day for 60 days (comparator arm). The primary outcome is the efficacy of three different BNZ therapeutic schemes in terms of dose and duration. Efficacy will be assessed according to the proportion of patients with sustained parasitic load suppression in peripheral blood measured by polymerase chain reaction. The secondary outcomes are related to pharmacokinetics and drug tolerability. The follow-up will be 12 months from randomization to end of study participation. Recruitment was started in April 2018. CONCLUSION This is a clinical trial conducted for the assessment of different dose schemes of BNZ compared with the standard treatment regimen for the treatment of CD in the chronic phase. MULTIBENZ may help to clarify which is the most adequate BNZ regimen in terms of efficacy and safety, predicated on sustained parasitic load suppression in peripheral blood. TRIAL REGISTRATION ClinicalTrials.gov, NCT03191162. Registered on 19 June 2017.
Collapse
Affiliation(s)
- D Molina-Morant
- Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Universitat Autònoma de Barcelona, P° Vall d'Hebron 119, Edifici Mediterrània, VHIR, 08035, Barcelona, Spain
| | - M L Fernández
- Departamento de Clínica, Patología y Tratamiento, Instituto Nacional de Parasitología Dr. Mario Fatala Chaben, Ministerio de Salud y Desarrollo Social, Buenos Aires, Argentina
| | - P Bosch-Nicolau
- Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Universitat Autònoma de Barcelona, P° Vall d'Hebron 119, Edifici Mediterrània, VHIR, 08035, Barcelona, Spain
| | - E Sulleiro
- Microbiology Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M Bangher
- Instituto de Cardiología de Corrientes Juana Francisca Cabral (Argentina), Corrientes, Argentina
| | - F Salvador
- Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Universitat Autònoma de Barcelona, P° Vall d'Hebron 119, Edifici Mediterrània, VHIR, 08035, Barcelona, Spain
| | - A Sanchez-Montalva
- Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Universitat Autònoma de Barcelona, P° Vall d'Hebron 119, Edifici Mediterrània, VHIR, 08035, Barcelona, Spain
| | - A L P Ribeiro
- Programa de Pós-graduação Infectologia e Medicina Tropical, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - A M B de Paula
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, MG, Brazil
| | - S Eloi
- Programa de Pós-graduação em Patologia, Departamento de Propedêutica Complementar, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Faculdade de Medicina da Universidade José do Rosário Vellano, Belo Horizonte, Brazil
| | - R Correa-Oliveira
- René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Brazil
| | - J C Villar
- Faculty of Health Sciences, Universidad Autónoma de Bucaramanga and Research Department, Bucaramanga, Colombia
- Fundación Cardioinfantil - Instituto de Cardiología, Bogotá, Colombia
| | - S Sosa-Estani
- Chagas Clinical Program, Drugs for Neglected Disease initiative (DNDi), Geneva, Switzerland
- Epidemiology and Public Health Research Center, CONICET, Buenos Aires, Argentina
| | - I Molina
- Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Universitat Autònoma de Barcelona, P° Vall d'Hebron 119, Edifici Mediterrània, VHIR, 08035, Barcelona, Spain.
| |
Collapse
|
60
|
Altamura F, Rajesh R, Catta-Preta CMC, Moretti NS, Cestari I. The current drug discovery landscape for trypanosomiasis and leishmaniasis: Challenges and strategies to identify drug targets. Drug Dev Res 2020; 83:225-252. [PMID: 32249457 DOI: 10.1002/ddr.21664] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/05/2020] [Accepted: 03/13/2020] [Indexed: 12/11/2022]
Abstract
Human trypanosomiasis and leishmaniasis are vector-borne neglected tropical diseases caused by infection with the protozoan parasites Trypanosoma spp. and Leishmania spp., respectively. Once restricted to endemic areas, these diseases are now distributed worldwide due to human migration, climate change, and anthropogenic disturbance, causing significant health and economic burden globally. The current chemotherapy used to treat these diseases has limited efficacy, and drug resistance is spreading. Hence, new drugs are urgently needed. Phenotypic compound screenings have prevailed as the leading method to discover new drug candidates against these diseases. However, the publication of the complete genome sequences of multiple strains, advances in the application of CRISPR/Cas9 technology, and in vivo bioluminescence-based imaging have set the stage for advancing target-based drug discovery. This review analyses the limitations of the narrow pool of available drugs presently used for treating these diseases. It describes the current drug-based clinical trials highlighting the most promising leads. Furthermore, the review presents a focused discussion on the most important biological and pharmacological challenges that target-based drug discovery programs must overcome to advance drug candidates. Finally, it examines the advantages and limitations of modern research tools designed to identify and validate essential genes as drug targets, including genomic editing applications and in vivo imaging.
Collapse
Affiliation(s)
- Fernando Altamura
- Institute of Parasitology, McGill University, Ste Anne de Bellevue, Quebec, Canada
| | - Rishi Rajesh
- Institute of Parasitology, McGill University, Ste Anne de Bellevue, Quebec, Canada
| | | | - Nilmar S Moretti
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Igor Cestari
- Institute of Parasitology, McGill University, Ste Anne de Bellevue, Quebec, Canada
| |
Collapse
|
61
|
Varghese S, Rahmani R, Russell S, Deora GS, Ferrins L, Toynton A, Jones A, Sykes M, Kessler A, Eufrásio A, Cordeiro AT, Sherman J, Rodriguez A, Avery VM, Piggott MJ, Baell JB. Discovery of Potent N-Ethylurea Pyrazole Derivatives as Dual Inhibitors of Trypanosoma brucei and Trypanosoma cruzi. ACS Med Chem Lett 2020; 11:278-285. [PMID: 32184957 DOI: 10.1021/acsmedchemlett.9b00218] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/09/2019] [Indexed: 12/21/2022] Open
Abstract
Trypanosoma brucei (T. brucei) and Trypanosoma cruzi (T. cruzi) are causative agents of parasitic diseases known as human African trypanosomiasis and Chagas disease, respectively. Together, these diseases affect 68 million people around the world. Current treatments are unsatisfactory, frequently associated with intolerable side-effects, and generally inadequate in treating all stages of disease. In this paper, we report the discovery of N-ethylurea pyrazoles that potently and selectively inhibit the viability of T. brucei and T. cruzi. Sharp and logical SAR led to the identification of 54 as the best compound, with an in vitro IC50 of 9 nM and 16 nM against T. b. brucei and T. cruzi, respectively. Compound 54 demonstrates favorable physicochemical properties and was efficacious in a murine model of Chagas disease, leading to undetectable parasitemia within 6 days when CYP metabolism was inhibited.
Collapse
Affiliation(s)
- Swapna Varghese
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Raphaël Rahmani
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Stephanie Russell
- Chemistry, School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, Perth, Western Australia 6009, Australia
| | - Girdhar Singh Deora
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lori Ferrins
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Arthur Toynton
- Chemistry, School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, Perth, Western Australia 6009, Australia
| | - Amy Jones
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
| | - Melissa Sykes
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
| | | | - Amanda Eufrásio
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, São Paulo 13083-970, Brazil
| | - Artur Torres Cordeiro
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, São Paulo 13083-970, Brazil
| | - Julian Sherman
- Anti-Infectives Screening Core, New York University School of Medicine, New York 10016, United States
| | - Ana Rodriguez
- Anti-Infectives Screening Core, New York University School of Medicine, New York 10016, United States
| | - Vicky M. Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
| | - Matthew J. Piggott
- Chemistry, School of Molecular Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, Perth, Western Australia 6009, Australia
| | - Jonathan B. Baell
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
- ARC Centre for Fragment-Based Design, Monash University, Parkville, Victoria 3052, Australia
- Australian Translational Medicinal Chemistry Facility (ATMCF), Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
62
|
Sykes ML, Hilko DH, Kung LI, Poulsen SA, Avery VM. Investigation of pyrimidine nucleoside analogues as chemical probes to assess compound effects on the proliferation of Trypanosoma cruzi intracellular parasites. PLoS Negl Trop Dis 2020; 14:e0008068. [PMID: 32163414 PMCID: PMC7112222 DOI: 10.1371/journal.pntd.0008068] [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: 06/16/2019] [Revised: 04/01/2020] [Accepted: 01/15/2020] [Indexed: 02/05/2023] Open
Abstract
Trypanosoma cruzi parasites utilise de novo pyrimidine biosynthesis to produce DNA and survive within mammalian host cells. This pathway can be hijacked to assess the replication of intracellular parasites with the exogenous addition of a DNA specific probe. To identify suitable probe compounds for this application, a collection of pyrimidine nucleoside analogues was assessed for incorporation into T. cruzi intracellular amastigote DNA using image-based technology and script-based analysis. Associated mammalian cell toxicity of these compounds was also determined against both the parasite host cells (3T3 cells) and HEK293 cells. Incorporation of 5-ethynyl-2′-deoxyuridine (EdU) into parasite DNA was the most effective of the probes tested, with minimal growth inhibition observed following either two or four hours EdU exposure. EdU was subsequently utilised as a DNA probe, followed by visualisation with click chemistry to a fluorescent azide, to assess the impact of drugs and compounds with previously demonstrated activity against T. cruzi parasites, on parasite replication. The inhibitory profiles of these molecules highlight the benefit of this approach for identifying surviving parasites post-treatment in vitro and classifying compounds as either fast or slow-acting. F-ara-EdU resulted in <50% activity observed against T. cruzi amastigotes following 48 hours incubation, at 73 μM. Collectively, this supports the further development of pyrimidine nucleosides as chemical probes to investigate replication of the parasite T. cruzi. Chagas disease occurs within 21 countries in the Americas, causes over 10, 000 deaths per year and a further 25 million people are at risk of being infected. The cause of Chagas disease is Trypanosoma cruzi, a single celled protozoan parasite, which enters the bloodstream of a host by the bite of a “kissing bug”. In advanced disease stages, the parasite hides in heart and gut tissue and is difficult to treat. Identifying the replicative ability of these parasites is important to understanding Chagas disease progression and the effectiveness of compounds and drugs for treatment. By testing a panel of nucleoside analogues that may incorporate into DNA during synthesis, we developed an image-based method with a fluorescently-labelled DNA probe to identify replicating parasites. This method has effectively shown that drugs used to treat the parasite are able to clear intracellular infection, whilst a compound that was not efficacious in clinical trials leaves replicating T. cruzi behind. This methodology can be used to understand the action of further compounds and supports the identification of new, less toxic probes to assess intracellular parasite replication.
Collapse
Affiliation(s)
- Melissa Louise Sykes
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| | - David Hugh Hilko
- Chemical Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| | - Livia Isabella Kung
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia.,Institute of Molecular Health Sciences, ETH Zurich, Switzerland
| | - Sally-Ann Poulsen
- Chemical Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| | - Vicky Marie Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia
| |
Collapse
|
63
|
Abstract
About one-sixth of the world's population is affected by a neglected tropical disease as defined by the World Health Organization and Center for Disease Control. Parasitic diseases comprise most of the neglected tropical disease list and they are causing enormous amounts of disability, morbidity, mortality, and healthcare costs worldwide. The burden of disease of the top five parasitic diseases has been estimated to amount to a total 23 million disability-adjusted life-years. Despite the massive health and economic impact, most drugs currently used for the treatment of parasitic diseases have been developed decades ago and insufficient novel drugs are being developed. The current review provides a compilation of the systemic and target-site pharmacokinetics of established antiparasitic drugs. Knowledge of the pharmacokinetic profile of drugs allows for the examination and possibly optimization of existing dosing schemes. Many symptoms of parasitic diseases are caused by parasites residing in different host tissues. Penetration of the antiparasitic drug into these tissues, the target site of infection, is a prerequisite for a successful treatment of the disease. Therefore, for the examination and improvement of established dosing regimens, not only the plasma but also the tissue pharmacokinetics of the drug have to be considered. For the current paper, almost 7000 scientific articles were identified and screened from which 429 were reviewed in detail and 100 were included in this paper. Systemic pharmacokinetics are available for most antiparasitic drugs but in many cases, not for all the relevant patient populations and only for single- or multiple-dose administration. Systemic pharmacokinetic data in patients with organ impairment and target-site pharmacokinetic data for relevant tissues and body fluids are mostly lacking. To improve the treatment of patients with parasitic diseases, research in these areas is urgently needed.
Collapse
Affiliation(s)
- Valentin Al Jalali
- Department of Clinical Pharmacology, Vienna University Hospital, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Vienna University Hospital, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
64
|
Santos SS, de Araújo RV, Giarolla J, Seoud OE, Ferreira EI. Searching for drugs for Chagas disease, leishmaniasis and schistosomiasis: a review. Int J Antimicrob Agents 2020; 55:105906. [PMID: 31987883 DOI: 10.1016/j.ijantimicag.2020.105906] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/16/2022]
Abstract
Chagas disease, leishmaniasis and schistosomiasis are neglected diseases (NDs) and are a considerable global challenge. Despite the huge number of people infected, NDs do not create interest from pharmaceutical companies because the associated revenue is generally low. Most of the research on these diseases has been conducted in academic institutions. The chemotherapeutic armamentarium for NDs is scarce and inefficient and better drugs are needed. Researchers have found some promising potential drug candidates using medicinal chemistry and computational approaches. Most of these compounds are synthetic but some are from natural sources or are semi-synthetic. Drug repurposing or repositioning has also been greatly stimulated for NDs. This review considers some potential drug candidates and provides details of their design, discovery and activity.
Collapse
Affiliation(s)
- Soraya Silva Santos
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Renan Vinicius de Araújo
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Jeanine Giarolla
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Omar El Seoud
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil
| | - Elizabeth Igne Ferreira
- Laboratory of Design and Synthesis of Chemotherapeutics Potentially Active in Neglected Diseases (LAPEN), Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo-USP, Avenue Professor Lineu Prestes, 580-Building 13, São Paulo SP, 05508-900, Brazil.
| |
Collapse
|
65
|
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: 8] [Impact Index Per Article: 1.6] [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
|
66
|
Borsari C, Jiménez-Antón MD, Eick J, Bifeld E, Torrado JJ, Olías-Molero AI, Corral MJ, Santarem N, Baptista C, Severi L, Gul S, Wolf M, Kuzikov M, Ellinger B, Reinshagen J, Witt G, Linciano P, Tait A, Costantino L, Luciani R, Tejera Nevado P, Zander-Dinse D, Franco CH, Ferrari S, Moraes CB, Cordeiro-da-Silva A, Ponterini G, Clos J, Alunda JM, Costi MP. Discovery of a benzothiophene-flavonol halting miltefosine and antimonial drug resistance in Leishmania parasites through the application of medicinal chemistry, screening and genomics. Eur J Med Chem 2019; 183:111676. [DOI: 10.1016/j.ejmech.2019.111676] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/01/2019] [Accepted: 09/02/2019] [Indexed: 01/24/2023]
|
67
|
Villalta F, Rachakonda G. Advances in preclinical approaches to Chagas disease drug discovery. Expert Opin Drug Discov 2019; 14:1161-1174. [PMID: 31411084 PMCID: PMC6779130 DOI: 10.1080/17460441.2019.1652593] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/02/2019] [Indexed: 12/21/2022]
Abstract
Introduction: Chagas disease affects 8-10 million people worldwide, mainly in Latin America. The current therapy for Chagas disease is limited to nifurtimox and benznidazole, which are effective in treating only the acute phase of the disease but with severe side effects. Therefore, there is an unmet need for new drugs and for the exploration of innovative approaches which may lead to the discovery of new effective and safe drugs for its treatment. Areas covered: The authors report and discuss recent approaches including structure-based design that have led to the discovery of new promising small molecule candidates for Chagas disease which affect prime targets that intervene in the sterol pathway of T. cruzi. Other trypanosome targets, phenotypic screening, the use of artificial intelligence and the challenges with Chagas disease drug discovery are also discussed. Expert opinion: The application of recent scientific innovations to the field of Chagas disease have led to the discovery of new promising drug candidates for Chagas disease. Phenotypic screening brought new hits and opportunities for drug discovery. Artificial intelligence also has the potential to accelerate drug discovery in Chagas disease and further research into this is warranted.
Collapse
Affiliation(s)
- Fernando Villalta
- Department of Microbiology, Immunology and Physiology, School of Medicine, Meharry Medical College , Nashville , TN , USA
| | - Girish Rachakonda
- Department of Microbiology, Immunology and Physiology, School of Medicine, Meharry Medical College , Nashville , TN , USA
| |
Collapse
|
68
|
In Vitro Benznidazole and Nifurtimox Susceptibility Profile of Trypanosoma cruzi Strains Belonging to Discrete Typing Units TcI, TcII, and TcV. Pathogens 2019; 8:pathogens8040197. [PMID: 31635071 PMCID: PMC6963282 DOI: 10.3390/pathogens8040197] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 01/06/2023] Open
Abstract
We ascertain the in vitro Benznidazole (BZN) and Nifurtimox (NFX) susceptibility pattern of epimastigotes, trypomastigotes, and amastigotes of 21 T. cruzi strains, from patients, reservoir, and triatomine bugs of various geographic origins. Using this panel of isolates, we compute the Epidemiological cut off value (COwt). Then, the frequency of the susceptible phenotype (Wild type) towards benznidazole (BZN) and nifurtimox (NFX) within this set of strains belonging to three discrete typing units (DTUs), TcI, TcII, and TcV, was deduced. We observed that the susceptibility status of individual T. cruzi isolates toward BZN and NFX is related to the genetic background and underlying factors that are probably related to the individual life trait history of each strain. Analyzing drug susceptibility in this conceptual framework would offer the possibility to evidence a link between isolates expressing a low susceptibility level (not wild-type) as defined by the COwt value and none-curative treatment. It will also permit us to track drug-resistant parasites in the T. cruzi population.
Collapse
|
69
|
Synthesis and structure-activity relationship of nitrile-based cruzain inhibitors incorporating a trifluoroethylamine-based P2 amide replacement. Bioorg Med Chem 2019; 27:115083. [PMID: 31561938 DOI: 10.1016/j.bmc.2019.115083] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/22/2019] [Accepted: 08/26/2019] [Indexed: 02/02/2023]
Abstract
The structure-activity relationship for nitrile-based cruzain inhibitors incorporating a P2 amide replacement based on trifluoroethylamine was explored by deconstruction of a published series of inhibitors. It was demonstrated that the P3 biphenyl substituent present in the published inhibitor structures could be truncated to phenyl with only a small loss of affinity. The effects of inverting the configuration of the P2 amide replacement and linking a benzyl substituent at P1 were observed to be strongly nonadditive. We show that plotting affinity against molecular size provides a means to visualize both the molecular size efficiency of structural transformations and the nonadditivity in the structure-activity relationship. We also show how the relationship between affinity and lipophilicity, measured by high-performance liquid chromatography with an immobilized artificial membrane stationary phase, may be used to normalize affinity with respect to lipophilicity.
Collapse
|
70
|
Silva ACC, Brelaz-de-Castro MCA, Leite ACL, Pereira VRA, Hernandes MZ. Chagas Disease Treatment and Rational Drug Discovery: A Challenge That Remains. Front Pharmacol 2019; 10:873. [PMID: 31427977 PMCID: PMC6690016 DOI: 10.3389/fphar.2019.00873] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/08/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ana Catarina Cristovão Silva
- Laboratório de Imunopatologia e Biologia Molecular, Departamento de Imunologia, Instituto Aggeu Magalhães, Recife, Brazil.,Programa de Pós-graduação em Inovação Terapêutica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Brazil
| | - Maria Carolina Accioly Brelaz-de-Castro
- Laboratório de Imunopatologia e Biologia Molecular, Departamento de Imunologia, Instituto Aggeu Magalhães, Recife, Brazil.,Laboratório de Parasitologia, Centro Acadêmico de Vitória, Universidade Federal de Pernambuco, Vitória de Santo Antão, Brazil
| | | | - Valéria Rêgo Alves Pereira
- Laboratório de Imunopatologia e Biologia Molecular, Departamento de Imunologia, Instituto Aggeu Magalhães, Recife, Brazil
| | | |
Collapse
|
71
|
Cianni L, Feldmann CW, Gilberg E, Gütschow M, Juliano L, Leitão A, Bajorath J, Montanari CA. Can Cysteine Protease Cross-Class Inhibitors Achieve Selectivity? J Med Chem 2019; 62:10497-10525. [DOI: 10.1021/acs.jmedchem.9b00683] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Lorenzo Cianni
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense, 400, 23566-590 São Carlos, SP, Brazil
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Christian Wolfgang Feldmann
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Erik Gilberg
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Michael Gütschow
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Luiz Juliano
- A. C. Camargo Cancer Center and São Paulo Medical School of Federal University of São Paulo, Rua Professor Antônio Prudente, 211, 01509-010 São Paulo, SP, Brazil
| | - Andrei Leitão
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense, 400, 23566-590 São Carlos, SP, Brazil
| | - Jürgen Bajorath
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Endenicher Allee 19c, D-53115 Bonn, Germany
| | - Carlos A. Montanari
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense, 400, 23566-590 São Carlos, SP, Brazil
| |
Collapse
|
72
|
Salas-Sarduy E, Niemirowicz GT, José Cazzulo J, Alvarez VE. Target-based Screening of the Chagas Box: Setting Up Enzymatic Assays to Discover Specific Inhibitors Across Bioactive Compounds. Curr Med Chem 2019; 26:6672-6686. [PMID: 31284853 DOI: 10.2174/0929867326666190705160637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/10/2018] [Accepted: 11/07/2018] [Indexed: 11/22/2022]
Abstract
Chagas disease is a neglected tropical illness caused by the protozoan parasite Trypanosoma cruzi. The disease is endemic in Latin America with about 6 million people infected and many more being at risk. Only two drugs are available for treatment, Nifurtimox and Benznidazole, but they have a number of side effects and are not effective in all cases. This makes urgently necessary the development of new drugs, more efficient, less toxic and affordable to the poor people, who are most of the infected population. In this review we will summarize the current strategies used for drug discovery considering drug repositioning, phenotyping screenings and target-based approaches. In addition, we will describe in detail the considerations for setting up robust enzymatic assays aimed at identifying and validating small molecule inhibitors in high throughput screenings.
Collapse
Affiliation(s)
- Emir Salas-Sarduy
- Instituto de Investigaciones Biotecnologicas Dr. Rodolfo A. Ugalde - Instituto Tecnologico de Chascomus (IIB-INTECH), Universidad Nacional de San Martin (UNSAM) - Consejo Nacional de Investigaciones Cientificas y Técnicas (CONICET), Campus Miguelete, Av. 25 de Mayo y Francia, 1650 San Martin, Buenos Aires, Argentina
| | - Gabriela T Niemirowicz
- Instituto de Investigaciones Biotecnologicas Dr. Rodolfo A. Ugalde - Instituto Tecnologico de Chascomus (IIB-INTECH), Universidad Nacional de San Martin (UNSAM) - Consejo Nacional de Investigaciones Cientificas y Técnicas (CONICET), Campus Miguelete, Av. 25 de Mayo y Francia, 1650 San Martin, Buenos Aires, Argentina
| | - Juan José Cazzulo
- Instituto de Investigaciones Biotecnologicas Dr. Rodolfo A. Ugalde - Instituto Tecnologico de Chascomus (IIB-INTECH), Universidad Nacional de San Martin (UNSAM) - Consejo Nacional de Investigaciones Cientificas y Técnicas (CONICET), Campus Miguelete, Av. 25 de Mayo y Francia, 1650 San Martin, Buenos Aires, Argentina
| | - Vanina E Alvarez
- Instituto de Investigaciones Biotecnologicas Dr. Rodolfo A. Ugalde - Instituto Tecnologico de Chascomus (IIB-INTECH), Universidad Nacional de San Martin (UNSAM) - Consejo Nacional de Investigaciones Cientificas y Técnicas (CONICET), Campus Miguelete, Av. 25 de Mayo y Francia, 1650 San Martin, Buenos Aires, Argentina
| |
Collapse
|
73
|
Synergic Effect of Allopurinol in Combination with Nitroheterocyclic Compounds against Trypanosoma cruzi. Antimicrob Agents Chemother 2019; 63:AAC.02264-18. [PMID: 30962342 PMCID: PMC6535576 DOI: 10.1128/aac.02264-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/24/2019] [Indexed: 11/20/2022] Open
Abstract
Combination therapy has gained attention as a possible strategy for overcoming the limitations of the present therapeutic arsenal for Chagas disease. The aim of this study was to evaluate the effect of allopurinol in association with nitroheterocyclic compounds on infection with the Y strain of Trypanosoma cruzi The in vitro effect of allopurinol plus benznidazole or nifurtimox on intracellular amastigotes in infected H9c2 cells was assessed in a 72-h assay. The interactions were classified as synergic for both allopurinol-nifurtimox (sums of fractional inhibitory concentrations [∑FICs] = 0.49 ± 0.08) and allopurinol-benznidazole (∑FICs = 0.48 ± 0.09). In the next step, infected Swiss mice were treated with allopurinol at 30, 60, and 90 mg/kg of body weight and with benznidazole at 25, 50, and 75 mg/kg in monotherapy and in combination at the same doses; as a reference treatment, another group of animals received benznidazole at 100 mg/kg. Allopurinol in monotherapy led to a smaller or nil effect in the reduction of parasite load and mortality rate. Treatment with benznidazole at suboptimal doses induced a transient suppression of parasitaemia with subsequent relapse in all animals treated with 25 and 50 mg/kg and in 80% of those that received 75 mg/kg. Administration of the drugs in combination significantly increased the cure rate to 60 to 100% among mice treated with benznidazole at 75 mg/kg plus 30, 60, or 90 mg/kg of allopurinol. These results show a positive interaction between allopurinol and benznidazole, and since both drugs are commercially available, their use in combination may be considered for the assessment in the treatment of Chagas disease patients.
Collapse
|
74
|
Roquero I, Cantizani J, Cotillo I, Manzano MP, Kessler A, Martín JJ, McNamara CW. Novel chemical starting points for drug discovery in leishmaniasis and Chagas disease. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2019; 10:58-68. [PMID: 31158574 PMCID: PMC6545338 DOI: 10.1016/j.ijpddr.2019.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 05/13/2019] [Accepted: 05/16/2019] [Indexed: 11/25/2022]
Abstract
Visceral leishmaniasis (VL) and Chagas disease (CD) are caused by kinetoplastid parasites that affect millions of people worldwide and impart a heavy burden against human health. Due to the partial efficacy and toxicity-related limitations of the existing treatments, there is an urgent need to develop novel therapies with superior efficacy and safety profiles to successfully treat these diseases. Herein we report the application of whole-cell phenotypic assays to screen a set of 150,000 compounds against Leishmania donovani, a causative agent of VL, and Trypanosoma cruzi, the causative agent of CD, with the objective of finding new starting points to develop novel drugs to effectively treat and control these diseases. The screening campaign, conducted with the purpose of global open access, identified twelve novel chemotypes with low to sub-micromolar activity against T. cruzi and/or L. donovani. We disclose these hit structures and associated activity with the goal to contribute to the drug discovery community by providing unique chemical tools to probe kinetoplastid biology and as hit-to-lead candidates for drug discovery. An open source drug discovery screen between The Tres Cantos Open Lab Foundation and Calibr at Scripps Research. High-throughput phenotypic screen of a 150,000-compound library against T. cruzi and L. donovani. Identification and characterization of 12 novel chemical series. 7 of those 12 chemical series are active against both T. cruzi and L. donovani. These chemical series may be valuable tools to identify new drug targets.
Collapse
Affiliation(s)
- Irene Roquero
- Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus, GSK, Tres Cantos, Spain
| | - Juan Cantizani
- Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus, GSK, Tres Cantos, Spain
| | - Ignacio Cotillo
- Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus, GSK, Tres Cantos, Spain
| | - M Pilar Manzano
- Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus, GSK, Tres Cantos, Spain
| | - Albane Kessler
- Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus, GSK, Tres Cantos, Spain
| | - J Julio Martín
- Diseases of the Developing World (DDW), Tres Cantos Medicines Development Campus, GSK, Tres Cantos, Spain.
| | | |
Collapse
|
75
|
Franco CH, Alcântara LM, Chatelain E, Freitas-Junior L, Moraes CB. Drug Discovery for Chagas Disease: Impact of Different Host Cell Lines on Assay Performance and Hit Compound Selection. Trop Med Infect Dis 2019; 4:E82. [PMID: 31108888 PMCID: PMC6630705 DOI: 10.3390/tropicalmed4020082] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/12/2019] [Accepted: 05/14/2019] [Indexed: 01/01/2023] Open
Abstract
Cell-based screening has become the major compound interrogation strategy in Chagas disease drug discovery. Several different cell lines have been deployed as host cells in screening assays. However, host cell characteristics and host-parasite interactions may play an important role when assessing anti-T. cruzi compound activity, ultimately impacting on hit discovery. To verify this hypothesis, four distinct mammalian cell lines (U2OS, THP-1, Vero and L6) were used as T. cruzi host cells in High Content Screening assays. Rates of infection varied greatly between different host cells. Susceptibility to benznidazole also varied, depending on the host cell and parasite strain. A library of 1,280 compounds was screened against the four different cell lines infected with T. cruzi, resulting in the selection of a total of 82 distinct compounds as hits. From these, only two hits were common to all four cell lines assays (2.4%) and 51 were exclusively selected from a single assay (62.2%). Infected U2OS cells were the most sensitive assay, as 55 compounds in total were identified as hits; infected THP-1 yielded the lowest hit rates, with only 16 hit compounds. Of the selected hits, compound FPL64176 presented selective anti-T. cruzi activity and could serve as a starting point for the discovery of new anti-chagasic drugs.
Collapse
Affiliation(s)
- Caio Haddad Franco
- Brazilian Biosciences National Laboratory, National Centre for Research in Energy and Materials, Campinas, SP 13083-970, Brazil.
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil.
| | - Laura Maria Alcântara
- Brazilian Biosciences National Laboratory, National Centre for Research in Energy and Materials, Campinas, SP 13083-970, Brazil.
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil.
| | - Eric Chatelain
- Drugs for Neglected Diseases Initiative, 1202 Geneva, Switzerland.
| | - Lucio Freitas-Junior
- Brazilian Biosciences National Laboratory, National Centre for Research in Energy and Materials, Campinas, SP 13083-970, Brazil.
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil.
| | - Carolina Borsoi Moraes
- Brazilian Biosciences National Laboratory, National Centre for Research in Energy and Materials, Campinas, SP 13083-970, Brazil.
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil.
| |
Collapse
|
76
|
Linciano P, Pozzi C, Iacono LD, di Pisa F, Landi G, Bonucci A, Gul S, Kuzikov M, Ellinger B, Witt G, Santarem N, Baptista C, Franco C, Moraes CB, Müller W, Wittig U, Luciani R, Sesenna A, Quotadamo A, Ferrari S, Pöhner I, Cordeiro-da-Silva A, Mangani S, Costantino L, Costi MP. Enhancement of Benzothiazoles as Pteridine Reductase-1 Inhibitors for the Treatment of Trypanosomatidic Infections. J Med Chem 2019; 62:3989-4012. [DOI: 10.1021/acs.jmedchem.8b02021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Pasquale Linciano
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Cecilia Pozzi
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Lucia dello Iacono
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Flavio di Pisa
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Giacomo Landi
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Alessio Bonucci
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Nuno Santarem
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Catarina Baptista
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Caio Franco
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisaem Energia e Materiais (CNPEM), 13083-100 Campinas, São Paulo, Brazil
| | - Carolina B. Moraes
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisaem Energia e Materiais (CNPEM), 13083-100 Campinas, São Paulo, Brazil
| | | | | | - Rosaria Luciani
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Antony Sesenna
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Antonio Quotadamo
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Stefania Ferrari
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | | | - Anabela Cordeiro-da-Silva
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Stefano Mangani
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Luca Costantino
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Maria Paola Costi
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| |
Collapse
|
77
|
Guedes-da-Silva FH, Batista DDGJ, Da Silva CF, Pavão BP, Batista MM, Moreira OC, Souza LRQ, Britto C, Rachakonda G, Villalta F, Lepesheva GI, Soeiro MDNC. Successful Aspects of the Coadministration of Sterol 14α-Demethylase Inhibitor VFV and Benznidazole in Experimental Mouse Models of Chagas Disease Caused by the Drug-Resistant Strain of Trypanosoma cruzi. ACS Infect Dis 2019; 5:365-371. [PMID: 30625275 DOI: 10.1021/acsinfecdis.8b00253] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Up to now, no vaccines are available for Chagas disease, and the current therapy is largely unsatisfactory. Novel imidazole-based scaffolds of protozoan sterol 14α-demethylase (CYP51) inhibitors have demonstrated potent antiparasitic activity with no acute toxicity. Presently our aim was to investigate the effectiveness of the experimental 14α-demethylase inhibitor VFV in the mouse models of Trypanosoma cruzi infection using a naturally drug-resistant Colombiana strain, under monotherapy and in association with the reference drug, benznidazole (Bz). The treatment with VFV resulted in complete parasitemia suppression and 100% animal survival when administered orally (given in 10% DMSO plus 5% Arabic gum) at 25 mg/kg (bid) for 60 days. However, as parasite relapse was found using VFV alone under this treatment scheme, the coadministration of VFV with Bz was assayed giving simultaneously (for 60 days, bid) by oral route, under two different drug vehicles (10% DMSO plus 5% Gum Arabic with or without 3% Tween 80). All tested mice groups resulted in >99.9% of parasitemia decrease and 100% animal survival. qPCR analysis performed on cyclophosphamide immunosuppressed mice revealed that, although presenting lack of cure, VFV given as monotherapy was 14-fold more active than Bz, and the coadministration of Bz plus VFV (given simultaneously, using 10% DMSO plus 5% Gum Arabic as vehicle) resulted in 106-fold lower blood parasitism as compared to the monotherapy of Bz. Another interesting finding was the parasitological cure in 70% of the animals treated with Bz and VFV when the coadministration was given using the VFV suspension in 10% DMSO + Arabic gum + Tween 80 (a formulation that we have found to provide a better pharmacokinetics), even after immunosuppression using cyclophosphamide cycles, supporting the promising aspect of the drug coadministration in improving the efficacy of therapeutic arsenal against T. cruzi.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Girish Rachakonda
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, 1005 Dr. DB Todd Jr. Blvd., Nashville, Tennessee 37208, United States,
| | - Fernando Villalta
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, 1005 Dr. DB Todd Jr. Blvd., Nashville, Tennessee 37208, United States,
| | - Galina I. Lepesheva
- Department of Biochemistry School of Medicine, Institute for Global Health, Vanderbilt University, 622A RRB, 2200 Pierce Avenue, Nashville, Tennessee 37232, United States
| | | |
Collapse
|
78
|
Menna-Barreto RFS. Cell death pathways in pathogenic trypanosomatids: lessons of (over)kill. Cell Death Dis 2019; 10:93. [PMID: 30700697 PMCID: PMC6353990 DOI: 10.1038/s41419-019-1370-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 12/10/2018] [Accepted: 12/13/2018] [Indexed: 12/19/2022]
Abstract
Especially in tropical and developing countries, the clinically relevant protozoa Trypanosoma cruzi (Chagas disease), Trypanosoma brucei (sleeping sickness) and Leishmania species (leishmaniasis) stand out and infect millions of people worldwide leading to critical social-economic implications. Low-income populations are mainly affected by these three illnesses that are neglected by the pharmaceutical industry. Current anti-trypanosomatid drugs present variable efficacy with remarkable side effects that almost lead to treatment discontinuation, justifying a continuous search for alternative compounds that interfere with essential and specific parasite pathways. In this scenario, the triggering of trypanosomatid cell death machinery emerges as a promising approach, although the exact mechanisms involved in unicellular eukaryotes are still unclear as well as the controversial biological importance of programmed cell death (PCD). In this review, the mechanisms of autophagy, apoptosis-like cell death and necrosis found in pathogenic trypanosomatids are discussed, as well as their roles in successful infection. Based on the published genomic and proteomic maps, the panel of trypanosomatid cell death molecules was constructed under different experimental conditions. The lack of PCD molecular regulators and executioners in these parasites up to now has led to cell death being classified as an unregulated process or incidental necrosis, despite all morphological evidence published. In this context, the participation of metacaspases in PCD was also not described, and these proteases play a crucial role in proliferation and differentiation processes. On the other hand, autophagic phenotype has been described in trypanosomatids under a great variety of stress conditions (drugs, starvation, among others) suggesting that this process is involved in the turnover of damaged structures in the protozoa and is not a cell death pathway. Death mechanisms of pathogenic trypanosomatids may be involved in pathogenesis, and the identification of parasite-specific regulators could represent a rational and attractive alternative target for drug development for these neglected diseases.
Collapse
|
79
|
Veale CGL. Unpacking the Pathogen Box-An Open Source Tool for Fighting Neglected Tropical Disease. ChemMedChem 2019; 14:386-453. [PMID: 30614200 DOI: 10.1002/cmdc.201800755] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Indexed: 12/13/2022]
Abstract
The Pathogen Box is a 400-strong collection of drug-like compounds, selected for their potential against several of the world's most important neglected tropical diseases, including trypanosomiasis, leishmaniasis, cryptosporidiosis, toxoplasmosis, filariasis, schistosomiasis, dengue virus and trichuriasis, in addition to malaria and tuberculosis. This library represents an ensemble of numerous successful drug discovery programmes from around the globe, aimed at providing a powerful resource to stimulate open source drug discovery for diseases threatening the most vulnerable communities in the world. This review seeks to provide an in-depth analysis of the literature pertaining to the compounds in the Pathogen Box, including structure-activity relationship highlights, mechanisms of action, related compounds with reported activity against different diseases, and, where appropriate, discussion on the known and putative targets of compounds, thereby providing context and increasing the accessibility of the Pathogen Box to the drug discovery community.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
80
|
Usefulness of Serial Blood Sampling and PCR Replicates for Treatment Monitoring of Patients with Chronic Chagas Disease. Antimicrob Agents Chemother 2019; 63:AAC.01191-18. [PMID: 30509941 PMCID: PMC6355557 DOI: 10.1128/aac.01191-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/11/2018] [Indexed: 02/04/2023] Open
Abstract
This work evaluated a serial blood sampling procedure to enhance the sensitivity of duplex real-time quantitative PCR (qPCR) for baseline detection and quantification of parasitic loads and posttreatment identification of failure in the context of clinical trials for treatment of chronic Chagas disease, namely, DNDi-CH-E1224-001 (ClinicalTrials.gov registration no. NCT01489228) and the MSF-DNDi PCR Sampling Optimization Study (NCT01678599). This work evaluated a serial blood sampling procedure to enhance the sensitivity of duplex real-time quantitative PCR (qPCR) for baseline detection and quantification of parasitic loads and posttreatment identification of failure in the context of clinical trials for treatment of chronic Chagas disease, namely, DNDi-CH-E1224-001 (ClinicalTrials.gov registration no. NCT01489228) and the MSF-DNDi PCR Sampling Optimization Study (NCT01678599). Patients from Cochabamba (n = 294), Tarija (n = 257), and Aiquile (n = 220) were enrolled. Three serial blood samples were collected at each time point, and qPCR triplicates were tested for each sample. The first two samples were collected during the same day and the third one 7 days later. A patient was considered PCR positive if at least one qPCR replicate was detectable. Cumulative results of multiple samples and qPCR replicates enhanced the proportion of pretreatment sample positivity from 54.8% to 76.2%, 59.5% to 77.8%, and 73.5% to 90.2% in Cochabamba, Tarija, and Aiquile cohorts, respectively. This strategy increased the detection of treatment failure from 72.9% to 91.7%, 77.8% to 88.9%, and 42.9% to 69.1% for E1224 low-, short-, and high-dosage regimens, respectively, and from 4.6% to 15.9% and 9.5% to 32.1% for the benznidazole arm in the DNDi-CH-E1224-001 and MSF-DNDi studies, respectively. The addition of the third blood sample and third qPCR replicate in patients with nondetectable PCR results in the first two samples gave a small, non-statistically significant improvement in qPCR positivity. No change in clinical sensitivity was seen with a blood volume increase from 5 to 10 ml. The monitoring of patients treated with placebo in the DNDi-CH-E1224-001 trial revealed fluctuations in parasitic loads and occasionally nondetectable results. In conclusion, a serial sampling strategy enhanced PCR sensitivity to detecting treatment failure during follow-up and has the potential for improving recruitment capacity in Chagas disease trials, which require an initial positive qPCR result for patient admission.
Collapse
|
81
|
Melo-Filho CC, Braga RC, Muratov EN, Franco CH, Moraes CB, Freitas-Junior LH, Andrade CH. Discovery of new potent hits against intracellular Trypanosoma cruzi by QSAR-based virtual screening. Eur J Med Chem 2018; 163:649-659. [PMID: 30562700 DOI: 10.1016/j.ejmech.2018.11.062] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 11/21/2018] [Accepted: 11/23/2018] [Indexed: 12/17/2022]
Abstract
Chagas disease is a neglected tropical disease (NTD) caused by the protozoan parasite Trypanosoma cruzi and is primarily transmitted to humans by the feces of infected Triatominae insects during their blood meal. The disease affects 6-8 million people, mostly in Latin America countries, and kills more people in the region each year than any other parasite-born disease, including malaria. Moreover, patient numbers are currently increasing in non-endemic, developed countries, such as Australia, Japan, Canada, and the United States. The treatment is limited to one drug, benznidazole, which is only effective in the acute phase of the disease and is very toxic. Thus, there is an urgent need to develop new, safer, and effective drugs against the chronic phase of Chagas disease. Using a QSAR-based virtual screening followed by in vitro experimental evaluation, we report herein the identification of novel potent and selective hits against T. cruzi intracellular stage. We developed and validated binary QSAR models for prediction of anti-trypanosomal activity and cytotoxicity against mammalian cells using the best practices for QSAR modeling. These models were then used for virtual screening of a commercial database, leading to the identification of 39 virtual hits. Further in vitro assays showed that seven compounds were potent against intracellular T. cruzi at submicromolar concentrations (EC50 < 1 μM) and were very selective (SI > 30). Furthermore, other six compounds were also inside the hit criteria for Chagas disease, which presented activity at low micromolar concentrations (EC50 < 10 μM) against intracellular T. cruzi and were also selective (SI > 15). Moreover, we performed a multi-parameter analysis for the comparison of tested compounds regarding their balance between potency, selectivity, and predicted ADMET properties. In the next studies, the most promising compounds will be submitted to additional in vitro and in vivo assays in acute model of Chagas disease, and can be further optimized for the development of new promising drug candidates against this important yet neglected disease.
Collapse
Affiliation(s)
- Cleber C Melo-Filho
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmacia, Universidade Federal de Goiás - UFG, Rua 240, Qd.87, Goiania, GO, 74605-510, Brazil
| | - Rodolpho C Braga
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmacia, Universidade Federal de Goiás - UFG, Rua 240, Qd.87, Goiania, GO, 74605-510, Brazil
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA; Department of Chemical Technology, Odessa National Polytechnic University, 1. Shevchenko Ave., Odessa, 65000, Ukraine
| | - Caio Haddad Franco
- National Laboratory of Biosciences (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP, 13083-970, Brazil
| | - Carolina B Moraes
- National Laboratory of Biosciences (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP, 13083-970, Brazil; Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Lucio H Freitas-Junior
- National Laboratory of Biosciences (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP, 13083-970, Brazil; Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Carolina Horta Andrade
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculdade de Farmacia, Universidade Federal de Goiás - UFG, Rua 240, Qd.87, Goiania, GO, 74605-510, Brazil.
| |
Collapse
|
82
|
Rodrigues-dos-Santos Í, Melo MF, de Castro L, Hasslocher-Moreno AM, do Brasil PEAA, Silvestre de Sousa A, Britto C, Moreira OC. Exploring the parasite load and molecular diversity of Trypanosoma cruzi in patients with chronic Chagas disease from different regions of Brazil. PLoS Negl Trop Dis 2018; 12:e0006939. [PMID: 30418976 PMCID: PMC6258420 DOI: 10.1371/journal.pntd.0006939] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 11/26/2018] [Accepted: 10/23/2018] [Indexed: 11/29/2022] Open
Abstract
Chagas disease is still a major public health issue in many Latin American countries. One of the current major challenges is to find an association between Trypanosoma cruzi discrete typing units (DTUs) and clinical manifestations of the disease. In this study, we used a multilocus conventional PCR and quantitative real time PCR (qPCR) approaches to perform the molecular typing and parasite load quantification directly from blood specimens of 65 chronic Chagas disease patients. All patients were recruited at the same health center, but their place of birth were widely distributed in different geographic regions of Brazil. Of the 65 patients, 35 (53.8%) presented positive amplification by real time qPCR, being 20 (30.7%) with the clinical indeterminate form and 15 (23.1%) with the cardiac form of the disease. The parasite load median for all positive patients was 2.54 [1.43-11.14] parasite equivalents/mL (par. Eq./mL), with the load ranging from 0.12 to 153.66 par. Eq./mL. Noteworthy, the parasite load was significantly higher in patients over 70 years old (median 20.05 [18.29-86.86] par. Eq./mL). Using guanidine-EDTA blood samples spiked with reference T. cruzi strains, belonging to the six DTUs, it was possible to genotype the parasite up to 0.5 par. Eq./mL, with high specificity. Of the patients with positive qPCR, it was possible to identify the T. cruzi DTU in 28 patients (80%). For the remaining patients (20%), at least a partial result was obtained. Analysis of specimens showed prevalences of TcVI, TcII and mixed infection TcVI+TcII equal to 40%, 17.1% and 14.3%, respectively. In addition, two patients were infected by TcV, and one patient was coinfected by TcIII+TcVI, These last three patients were in stage A of chronic chagasic cardiomyopathy (CCC), and they were born at the Bahia State (northeast region of Brazil). When T. cruzi genotypes were compared with the parasite load, more elevated parasite loads were observed in patients infected by TcII in general (parasite load median of 7.56 par. Eq./mL) in comparison to patients infected by TcVI (median of 2.35 par. Eq./mL). However, while the frequency of CCC was 50% in patients infected by TcVI and TcV, only 16.7% of patients infected by TcII evolved to CCC. Taking together, our results contribute to update the epidemiological knowledge of T. cruzi DTUs in Brazil, and highlight the age of patient and infection by TcII as important features that lead to the observation of higher parasitemia levels.
Collapse
Affiliation(s)
| | - Myllena F. Melo
- Laboratório de Biologia Molecular e Doenças Endêmicas, IOC /Fiocruz, Rio de Janeiro, Brazil
| | - Liane de Castro
- Laboratório de Pesquisa Clínica em Doença de Chagas, INI/ Fiocruz, Rio de Janeiro, Brazil
| | | | | | | | - Constança Britto
- Laboratório de Biologia Molecular e Doenças Endêmicas, IOC /Fiocruz, Rio de Janeiro, Brazil
| | - Otacilio C. Moreira
- Laboratório de Biologia Molecular e Doenças Endêmicas, IOC /Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
83
|
Müller Kratz J, Garcia Bournissen F, Forsyth CJ, Sosa-Estani S. Clinical and pharmacological profile of benznidazole for treatment of Chagas disease. Expert Rev Clin Pharmacol 2018; 11:943-957. [DOI: 10.1080/17512433.2018.1509704] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Jadel Müller Kratz
- Chagas Clinical Program, Drugs for Neglected Disease initiative (DNDi), Geneva, Switzerland
| | - Facundo Garcia Bournissen
- Parasitology and Chagas Service, Buenos Aires Children Hospital Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Colin J. Forsyth
- Chagas Clinical Program, Drugs for Neglected Disease initiative (DNDi), Geneva, Switzerland
| | - Sergio Sosa-Estani
- Chagas Clinical Program, Drugs for Neglected Disease initiative (DNDi), Geneva, Switzerland
- Epidemiology and Public Health Research Center, CONICET, Buenos Aires, Argentina
| |
Collapse
|
84
|
Fonseca-Berzal C, Arán VJ, Escario JA, Gómez-Barrio A. Experimental models in Chagas disease: a review of the methodologies applied for screening compounds against Trypanosoma cruzi. Parasitol Res 2018; 117:3367-3380. [PMID: 30232605 DOI: 10.1007/s00436-018-6084-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/11/2018] [Indexed: 01/29/2023]
Abstract
One of the main problems of Chagas disease (CD), the parasitic infection caused by Trypanosoma cruzi, is the lack of a completely satisfactory treatment, which is currently based on two old nitroheterocyclic drugs (i.e., nifurtimox and benznidazole) that show important limitations for treating patients. In this context, many laboratories look for alternative therapies potentially applicable to the treatment, and therefore, research in CD chemotherapy works in the design of experimental protocols for detecting molecules with activity against T. cruzi. Phenotypic assays are considered the most valuable strategy for screening these antiparasitic compounds. Among them, in vitro experiments are the first step to test potential anti-T. cruzi drugs directly on the different parasite forms (i.e., epimastigotes, trypomastigotes, and amastigotes) and to detect cytotoxicity. Once the putative trypanocidal drug has been identified in vitro, it must be moved to in vivo models of T. cruzi infection, to explore (i) acute toxicity, (ii) efficacy during the acute infection, and (iii) efficacy in the chronic disease. Moreover, in silico approaches for predicting activity have emerged as a supporting tool for drug screening procedures. Accordingly, this work reviews those in vitro, in vivo, and in silico methods that have been routinely applied during the last decades, aiming to discover trypanocidal compounds that contribute to developing more effective CD treatments.
Collapse
Affiliation(s)
- Cristina Fonseca-Berzal
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Pza. Ramón y Cajal s/n, 28040, Madrid, Spain.
| | - Vicente J Arán
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), c/ Juan de la Cierva 3, 28006, Madrid, Spain
| | - José A Escario
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Pza. Ramón y Cajal s/n, 28040, Madrid, Spain
| | - Alicia Gómez-Barrio
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Pza. Ramón y Cajal s/n, 28040, Madrid, Spain
| |
Collapse
|
85
|
Quebrada Palacio LP, González MN, Hernandez-Vasquez Y, Perrone AE, Parodi-Talice A, Bua J, Postan M. Phenotypic diversity and drug susceptibility of Trypanosoma cruzi TcV clinical isolates. PLoS One 2018; 13:e0203462. [PMID: 30183775 PMCID: PMC6124804 DOI: 10.1371/journal.pone.0203462] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 08/21/2018] [Indexed: 12/24/2022] Open
Abstract
Trypanosoma cruzi is a genetically heterogeneous group of organisms that cause Chagas disease. It has been long suspected that the clinical outcome of the disease and response to therapeutic agents are, at least in part, related to the genetic characteristics of the parasite. Herein, we sought to validate the significance of the genotype of T. cruzi isolates recovered from patients with different clinical forms of Chagas disease living in Argentina on their biological behaviour and susceptibility to drugs. Genotype identification of the newly established isolates confirmed the reported predominance of TcV, with a minor frequency of TcI. Epimastigote sensitivity assays demonstrated marked dissimilar responses to benznidazole, nifurtimox, pentamidine and dihydroartemisinin in vitro. Two TcV isolates exhibiting divergent response to benznidazole in epimastigote assays were further tested for the expression of anti-oxidant proteins. Benznidazole-resistant BOL-FC10A epimastigotes had decreased expression of Old Yellow Enzyme and cytosolic superoxide dismutase, and overexpression of mitochondrial superoxide dismutase and tryparedoxin- 1, compared to benznidazole-susceptible AR-SE23C parasites. Drug sensitivity assays on intracellular amastigotes and trypomastigotes reproduced the higher susceptibility of AR-SE23C over BOL-FC10A parasites to benznidazole observed in epimastigotes assays. However, the susceptibility/resistance profile of amastigotes and trypomastigotes to nifurtimox, pentamidine and dihydroartemisinin varied markedly with respect to that of epimastigotes. C3H/He mice infected with AR-SE23C trypomastigotes had higher levels of parasitemia and mortality rate during the acute phase of infection compared to mice infected with BOL-FC10A trypomastigotes. Treatment of infected mice with benznidazole or nifurtimox was efficient to reduce patent parasitemia induced by either isolate. Nevertheless, qPCR performed at 70 dpi revealed parasite DNA in the blood of mice infected with AR-SE23C but not in BOL-FC10A infected mice. These results demonstrate high level of intra-type diversity which may represent an important obstacle for the testing of chemotherapeutic agents.
Collapse
Affiliation(s)
- Luz P. Quebrada Palacio
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina
| | - Mariela N. González
- Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina
| | - Yolanda Hernandez-Vasquez
- Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina
| | - Alina E. Perrone
- Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina
| | - Adriana Parodi-Talice
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Sección Genética, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Jacqueline Bua
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina
| | - Miriam Postan
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
86
|
Abstract
African trypanosomes cause lethal and neglected tropical diseases, known as sleeping sickness in humans and nagana in animals. Current therapies are limited, but fortunately, promising therapies are in advanced clinical and veterinary development, including acoziborole (AN5568 or SCYX-7158) and AN11736, respectively. These benzoxaboroles will likely be key to the World Health Organization's target of disease control by 2030. Their mode of action was previously unknown. We have developed a high-coverage overexpression library and use it here to explore drug mode of action in Trypanosoma brucei Initially, an inhibitor with a known target was used to select for drug resistance and to test massive parallel library screening and genome-wide mapping; this effectively identified the known target and validated the approach. Subsequently, the overexpression screening approach was used to identify the target of the benzoxaboroles, Cleavage and Polyadenylation Specificity Factor 3 (CPSF3, Tb927.4.1340). We validated the CPSF3 endonuclease as the target, using independent overexpression strains. Knockdown provided genetic validation of CPSF3 as essential, and GFP tagging confirmed the expected nuclear localization. Molecular docking and CRISPR-Cas9-based editing demonstrated how acoziborole can specifically block the active site and mRNA processing by parasite, but not host CPSF3. Thus, our findings provide both genetic and chemical validation for CPSF3 as an important drug target in trypanosomes and reveal inhibition of mRNA maturation as the mode of action of the trypanocidal benzoxaboroles. Understanding the mechanism of action of benzoxaborole-based therapies can assist development of improved therapies, as well as the prediction and monitoring of resistance, if or when it arises.
Collapse
|
87
|
Zingales B. Trypanosoma cruzi genetic diversity: Something new for something known about Chagas disease manifestations, serodiagnosis and drug sensitivity. Acta Trop 2018; 184:38-52. [PMID: 28941731 DOI: 10.1016/j.actatropica.2017.09.017] [Citation(s) in RCA: 263] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/18/2017] [Accepted: 09/18/2017] [Indexed: 11/27/2022]
Abstract
The genetic diversity of Trypanosoma cruzi, the protozoan agent of Chagas disease, is widely recognized. At present, T. cruzi is partitioned into seven discrete typing units (DTUs), TcI-TcVI and Tcbat. This article reviews the present knowledge on the parasite population structure, the evolutionary relationships among DTUs and their distinct, but not exclusive ecological and epidemiological associations. Different models for the origin of hybrid DTUs are examined, which agree that genetic exchange among T. cruzi populations is frequent and has contributed to the present parasite population structure. The geographic distribution of the prevalent DTUs in humans from the southern United States to Argentina is here presented and the circumstantial evidence of a possible association between T. cruzi genotype and Chagas disease manifestations is discussed. The available information suggests that parasite strains detected in patients, regardless of the clinical presentation, reflect the principal DTU circulating in the domestic transmission cycles of a particular region. In contrast, in several orally transmitted outbreaks, sylvatic strains are implicated. As a consequence of the genotypic and phenotypic differences of T. cruzi strains and the differential geographic distribution of DTUs in humans, regional variations in the sensitivity of the serological tests are verified. The natural resistance to benznidazole and nifurtimox, verified in vivo and in vitro for some parasite stocks, is not associated with any particular DTU, and does not explain the marked difference in the anti-parasitic efficacy of both drugs in the acute and chronic phases of Chagas disease. Throughout this review, it is emphasized that the interplay between parasite and host genetics should have an important role in the definition of Chagas disease pathogenesis, anti-T. cruzi immune response and chemotherapy outcome and should be considered in future investigations.
Collapse
Affiliation(s)
- Bianca Zingales
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Avenida Professor Lineu Prestes 748, 05508-000 São Paulo, SP, Brazil.
| |
Collapse
|
88
|
MacLean LM, Thomas J, Lewis MD, Cotillo I, Gray DW, De Rycker M. Development of Trypanosoma cruzi in vitro assays to identify compounds suitable for progression in Chagas' disease drug discovery. PLoS Negl Trop Dis 2018; 12:e0006612. [PMID: 30001347 PMCID: PMC6057682 DOI: 10.1371/journal.pntd.0006612] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/24/2018] [Accepted: 06/14/2018] [Indexed: 01/08/2023] Open
Abstract
Chagas' disease is responsible for significant mortality and morbidity in Latin America. Current treatments display variable efficacy and have adverse side effects, hence more effective, better tolerated drugs are needed. However, recent efforts have proved unsuccessful with failure of the ergosterol biosynthesis inhibitor posaconazole in phase II clinical trials despite promising in vitro and in vivo studies. The lack of translation between laboratory experiments and clinical outcome is a major issue for further drug discovery efforts. Our goal was to identify cell-based assays that could differentiate current nitro-aromatic drugs nifurtimox and benznidazole from posaconazole. Using a panel of T. cruzi strains including the six major lineages (TcI-VI), we found that strain PAH179 (TcV) was markedly less susceptible to posaconazole in vitro. Determination of parasite doubling and cycling times as well as EdU labelling experiments all indicate that this lack of sensitivity is due to the slow doubling and cycling time of strain PAH179. This is in accordance with ergosterol biosynthesis inhibition by posaconazole leading to critically low ergosterol levels only after multiple rounds of division, and is further supported by the lack of effect of posaconazole on the non-replicative trypomastigote form. A washout experiment with prolonged posaconazole treatment showed that, even for more rapidly replicating strains, this compound cannot clear all parasites, indicative of a heterogeneous parasite population in vitro and potentially the presence of quiescent parasites. Benznidazole in contrast was able to kill all parasites. The work presented here shows clear differentiation between the nitro-aromatic drugs and posaconazole in several assays, and suggests that in vitro there may be clinically relevant heterogeneity in the parasite population that can be revealed in long-term washout experiments. Based on these findings we have adjusted our in vitro screening cascade so that only the most promising compounds are progressed to in vivo experiments.
Collapse
Affiliation(s)
- Lorna M. MacLean
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
- * E-mail: (LML); (DWG)
| | - John Thomas
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Michael D. Lewis
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Ignacio Cotillo
- GlaxoSmithKline, Diseases of the Developing World, Tres Cantos, Madrid, Spain
| | - David W. Gray
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
- * E-mail: (LML); (DWG)
| | - Manu De Rycker
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
89
|
Outcome of E1224-Benznidazole Combination Treatment for Infection with a Multidrug-Resistant Trypanosoma cruzi Strain in Mice. Antimicrob Agents Chemother 2018; 62:AAC.00401-18. [PMID: 29555633 PMCID: PMC5971593 DOI: 10.1128/aac.00401-18] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 12/17/2022] Open
Abstract
Combination therapy has been proposed as an alternative therapeutic approach for the treatment of Chagas disease. In this study, we evaluated the effect of treatment with benznidazole combined with E1224 (ravuconazole prodrug) in an experimental murine model of acute infection. The first set of experiments assessed the range of E1224 doses required to induce parasitological cure using Trypanosoma cruzi strains with different susceptibilities to benznidazole (Y and Colombian). All E1224 doses were effective in suppressing the parasitemia and preventing death; however, parasitological cure was observed only in mice infected with Y strain. Considering these results, we evaluated the effect of combined treatment against Colombian, a multidrug-resistant T. cruzi strain. After exclusion of antagonistic effects using in vitro assays, infected mice were treated with E1224 and benznidazole in monotherapy or in combination at day 4 or 10 postinoculation. All treatments were well tolerated and effective in suppressing parasitemia; however, parasitological and PCR assays indicated no cure among mice treated with monotherapies. Intriguingly, the outcome of combination therapy was dependent on treatment onset. Early treatment using optimal doses of E1224-benznidazole induced a 100% cure rate, but this association could not eliminate a well-established infection. The beneficial effect of combination therapy was evidenced by further reductions of the patent parasitemia period in the group receiving combined therapy compared with monotherapies. Our results demonstrated a positive interaction between E1224 and benznidazole against murine T. cruzi infection using a multidrug-resistant strain and highlighted the importance of a stringent experimental model in the evaluation of new therapies.
Collapse
|
90
|
Kipandula W, Young SA, MacNeill SA, Smith TK. Screening of the MMV and GSK open access chemical boxes using a viability assay developed against the kinetoplastid Crithidia fasciculata. Mol Biochem Parasitol 2018; 222:61-69. [PMID: 29782894 DOI: 10.1016/j.molbiopara.2018.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/19/2018] [Accepted: 05/01/2018] [Indexed: 12/24/2022]
Abstract
Diseases caused by the pathogenic kinetoplastids continue to incapacitate and kill hundreds of thousands of people annually throughout the tropics and sub-tropics. Unfortunately, in the countries where these neglected diseases occur, financial obstacles to drug discovery and technical limitations associated with biochemical studies impede the development of new, safe, easy to administer and effective drugs. Here we report the development and optimisation of a Crithidia fasciculata resazurin viability assay, which is subsequently used for screening and identification of anti-crithidial compounds in the MMV and GSK open access chemical boxes. The screening assay had an average Z' factor of 0.7 and tolerated a maximum dimethyl sulfoxide concentration of up to 0.5%. We identified from multiple chemical boxes two compound series exhibiting nanomolar potency against C. fasciculata, one centred around a 5-nitrofuran-2-yl scaffold, a well-known moiety in several existing anti-infectives, and another involving a 2-(pyridin-2-yl) pyrimidin-4-amine scaffold which seems to have pan-kinetoplastid activity. This work facilitates the future use of C. fasciculata as a non-pathogenic and inexpensive biological resource to identify mode of action/protein target(s) of potentially pan-trypanocidal potent compounds. This knowledge will aid in the development of new treatments for African sleeping sickness, Chagas disease and leishmaniasis.
Collapse
Affiliation(s)
- Wakisa Kipandula
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews, Fife KY16 9ST, UK; Department of Medical Laboratory Sciences, College of Medicine, University of Malawi, Private Bag 360, Chichiri, Blantyre 3, Malawi
| | - Simon A Young
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews, Fife KY16 9ST, UK
| | - Stuart A MacNeill
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews, Fife KY16 9ST, UK
| | - Terry K Smith
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews, Fife KY16 9ST, UK.
| |
Collapse
|
91
|
Sánchez-Valdéz FJ, Padilla A, Wang W, Orr D, Tarleton RL. Spontaneous dormancy protects Trypanosoma cruzi during extended drug exposure. eLife 2018; 7:34039. [PMID: 29578409 PMCID: PMC5906098 DOI: 10.7554/elife.34039] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 03/25/2018] [Indexed: 12/19/2022] Open
Abstract
The ability of the Chagas disease agent Trypanosoma cruzi to resist extended in vivo exposure to highly effective trypanocidal compounds prompted us to explore the potential for dormancy and its contribution to failed drug treatments in this infection. We document the development of non-proliferating intracellular amastigotes in vivo and in vitro in the absence of drug treatment. Non-proliferative amastigotes ultimately converted to trypomastigotes and established infections in new host cells. Most significantly, dormant amastigotes were uniquely resistant to extended drug treatment in vivo and in vitro and could re-establish a flourishing infection after as many as 30 days of drug exposure. These results demonstrate a dormancy state in T. cruzi that accounts for the failure of highly cytotoxic compounds to completely resolve the infection. The ability of T. cruzi to establish dormancy throws into question current methods for identifying curative drugs but also suggests alternative therapeutic approaches. Chagas disease is one of the most harmful tropical diseases in the Americas. It affects millions of people, predominantly in Latin America. It is usually spread by kissing bugs infected with Trypanosoma cruzi parasites. It is considered a neglected tropical disease because few effective treatments and preventive methods are routinely used. Several drugs can kill T. cruzi parasites, but they often fail to cure the infection. Many people with Chagas disease go on to have life-long infections and eventually develop heart failure. The reason for the high rate of treatment failure is not known. It does not appear to be the result of the parasites developing resistance to the drugs. One possibility is that the parasites can hide in a dormant state in the body, dodging the toxic drugs and living to reproduce another day. Now, Sánchez-Valdéz et al. identify a dormant form of the T. cruzi parasite that allows the infection to persist after treatment. In the experiments, a non-reproducing form of the so-called amastigote stage of the T. cruzi parasite inside the host cells was observed in infected mice and human cells. While some of the amastigote parasites continue multiplying, a few stop even without drug treatment – but can resume multiplication at a later time. They may also be able to change into the trypomastigote form of the parasite, which can infect new cells. These non-multiplying amastigotes can survive drug treatment for as long as 30 days, whereas the multiplying amastigotes are killed by such drugs. However, the surviving amastigotes then reestablish active infections after treatment has stopped. The experiments explain why treatment so often fails to cure Chagas disease. This suggests new treatment strategies are needed, including using existing drugs for a longer time perhaps with less frequent doses. New therapies that kill the dormant amastigotes may also help. Treatments that overcome the parasite’s ability to hide, could stop the progression of the disease and prevent heart-related deaths in those with persistent T. cruzi infections.
Collapse
Affiliation(s)
| | - Angel Padilla
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, United States.,Department of Cellular Biology, University of Georgia, Athens, United States
| | - Wei Wang
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, United States
| | - Dylan Orr
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, United States
| | - Rick L Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, United States.,Department of Cellular Biology, University of Georgia, Athens, United States
| |
Collapse
|
92
|
Machado FC, Franco CH, Dos Santos Neto JV, Dias-Teixeira KL, Moraes CB, Lopes UG, Aktas BH, Schenkman S. Identification of di-substituted ureas that prevent growth of trypanosomes through inhibition of translation initiation. Sci Rep 2018; 8:4857. [PMID: 29559670 PMCID: PMC5861040 DOI: 10.1038/s41598-018-23259-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/08/2018] [Indexed: 01/25/2023] Open
Abstract
Some 1,3-diarylureas and 1-((1,4-trans)−4-aryloxycyclohexyl)−3-arylureas (cHAUs) activate heme-regulated kinase causing protein synthesis inhibition via phosphorylation of the eukaryotic translation initiation factor 2 (eIF2) in mammalian cancer cells. To evaluate if these agents have potential to inhibit trypanosome multiplication by also affecting the phosphorylation of eIF2 alpha subunit (eIF2α), we tested 25 analogs of 1,3-diarylureas and cHAUs against Trypanosoma cruzi, the agent of Chagas disease. One of them (I-17) presented selectivity close to 10-fold against the insect replicative forms and also inhibited the multiplication of T. cruzi inside mammalian cells with an EC50 of 1–3 µM and a selectivity of 17-fold. I-17 also prevented replication of African trypanosomes (Trypanosoma brucei bloodstream and procyclic forms) at similar doses. It caused changes in the T. cruzi morphology, arrested parasite cell cycle in G1 phase, and promoted phosphorylation of eIF2α with a robust decrease in ribosome association with mRNA. The activity against T. brucei also implicates eIF2α phosphorylation, as replacement of WT-eIF2α with a non-phosphorylatable eIF2α, or knocking down eIF2 protein kinase-3 by RNAi increased resistance to I-17. Therefore, we demonstrate that eIF2α phosphorylation can be engaged to develop trypanosome-static agents in general, and particularly by interfering with activity of eIF2 kinases.
Collapse
Affiliation(s)
- Fabricio Castro Machado
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, 04039-032, São Paulo, SP, Brazil
| | - Caio Haddad Franco
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, 04039-032, São Paulo, SP, Brazil.,Instituto Butantan, São Paulo, SP, Brazil
| | - Jose Vitorino Dos Santos Neto
- Laboratório de Parasitologia Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Karina Luiza Dias-Teixeira
- Laboratório de Parasitologia Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carolina Borsoi Moraes
- Instituto Butantan, São Paulo, SP, Brazil.,Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ulisses Gazos Lopes
- Laboratório de Parasitologia Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Bertal Huseyin Aktas
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA, 02115, United States.
| | - Sergio Schenkman
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, 04039-032, São Paulo, SP, Brazil.
| |
Collapse
|
93
|
3-pyridyl inhibitors with novel activity against Trypanosoma cruzi reveal in vitro profiles can aid prediction of putative cytochrome P450 inhibition. Sci Rep 2018; 8:4901. [PMID: 29559688 PMCID: PMC5861127 DOI: 10.1038/s41598-018-22043-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 02/09/2018] [Indexed: 01/04/2023] Open
Abstract
Using high throughput, high-content imaging, a proprietary library was screened against intracellular Trypanosoma cruzi amastigotes to identify compounds with novel activity against the parasite. Five inhibitors were discovered, which did not clear all of the parasites from 3T3 host cells following 48 hours exposure, and were identified as putative T. cruzi cytochrome P450 (TcCYP51) inhibitors. TcCYP51 inhibitors are not favourable for the drug discovery pipeline for treatment of Chagas Disease infection due to clinical and pre-clinical failures. To determine if there were in vitro inhibitory characteristics of these compounds that could aid the prediction of TcCYP51 inhibition further profiling using imaging and fluorescence based assays was undertaken. It was determined that in vitro profiles, coupled with analysis of chemical structure, could support the early prediction of putative TcCYP51 activity and thus enable early de-prioritisation of these compounds from progression through the drug discovery pipeline.
Collapse
|
94
|
Bacteria from Antarctic environments: diversity and detection of antimicrobial, antiproliferative, and antiparasitic activities. Polar Biol 2018. [DOI: 10.1007/s00300-018-2300-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
95
|
Searching for new drugs for Chagas diseases: triazole analogs display high in vitro activity against Trypanosoma cruzi and low toxicity toward mammalian cells. J Bioenerg Biomembr 2018; 50:81-91. [PMID: 29473131 DOI: 10.1007/s10863-018-9746-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/07/2018] [Indexed: 12/25/2022]
Abstract
Chagas disease is one of the most relevant endemic diseases in Latin America caused by the flagellate protozoan Trypanosoma cruzi. Nifurtimox and benzonidazole are the drugs used in the treatment of this disease, but they commonly are toxic and present severe side effects. New effective molecules, without collateral effects, has promoted the investigation to develop new lead compounds with to advance for clinical trials. Previously, 3-nitro-1H-1,2,4-triazole-based amines and 1,2,3-triazoles demonstrated significant trypanocidal activity against T. cruzi. In this paper, we synthesized a new series of 92 examples of 1,2,3-triazoles. Six compounds exhibited antiparasitic activity, 14, 25, 27, 31 and 40, 43 and were effective against epimastigotes of two strains of T. cruzi (Y and Dm28-C) and 25, 27 and 31 exhibited trypanocidal activity similar to benzonidazole. Notably, the compound 25 compared to benzonidazole increase the toxicity against T. cruzi, with no apparent toxicity to the cell line of mice macrophages or primary mice peritoneal macrophages. As results, we calculated selectivity indexes up to 2000 to 25 and 31 in both T. cruzi strains. Derivative 14 caused a trypanostatic effect because it did not damage external epimastigote membrane. Triazoles 40 and 43 impaired parasites viability using a pathway not dependent on ROS production.
Collapse
|
96
|
Uliassi E, Piazzi L, Belluti F, Mazzanti A, Kaiser M, Brun R, Moraes CB, Freitas-Junior LH, Gul S, Kuzikov M, Ellinger B, Borsari C, Costi MP, Bolognesi ML. Development of a Focused Library of Triazole-Linked Privileged-Structure-Based Conjugates Leading to the Discovery of Novel Phenotypic Hits against Protozoan Parasitic Infections. ChemMedChem 2018; 13:678-683. [PMID: 29451361 DOI: 10.1002/cmdc.201700786] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Indexed: 11/08/2022]
Abstract
Protozoan infections caused by Plasmodium, Leishmania, and Trypanosoma spp. contribute significantly to the burden of infectious diseases worldwide, causing severe morbidity and mortality. The inadequacy of available treatments calls for cost- and time-effective drug discovery endeavors. To this end, we envisaged the triazole linkage of privileged structures as an effective drug design strategy to generate a focused library of high-quality compounds. The versatility of this approach was combined with the feasibility of a phenotypic assay, integrated with early ADME-tox profiling. Thus, an 18-membered library was efficiently assembled via Huisgen cycloaddition of phenothiazine, biphenyl, and phenylpiperazine scaffolds. The resulting 18 compounds were then tested against seven parasite strains, and counter-screened for selectivity against two mammalian cell lines. In parallel, hERG and cytochrome P450 (CYP) inhibition, and mitochondrial toxicity were assessed. Remarkably, 10-((1-(3-([1,1'-biphenyl]-3-yloxy)propyl)-1H-1,2,3-triazol-5-yl)methyl)-10H-phenothiazine (7) and 10-(3-(1-(3-([1,1'-biphenyl]-3-yloxy)propyl)-1H-1,2,3-triazol-4-yl)propyl)-10H-phenothiazine (12) showed respective IC50 values of 1.8 and 1.9 μg mL-1 against T. cruzi, together with optimal selectivity. In particular, compound 7 showed a promising ADME-tox profile. Thus, hit 7 might be progressed as an antichagasic lead.
Collapse
Affiliation(s)
- Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Lorna Piazzi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Federica Belluti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Andrea Mazzanti
- Department of Industrial Chemistry "Toso Montonari", Alma Mater Studiorum-University of Bologna, Viale del Risorgimento 4, 40136, Bologna, Italy
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, 4002, Basel, Switzerland.,University of Basel, Petersplatz 1, 4003, Basel, Switzerland
| | - Reto Brun
- Swiss Tropical and Public Health Institute, 4002, Basel, Switzerland.,University of Basel, Petersplatz 1, 4003, Basel, Switzerland
| | - Carolina B Moraes
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), 13083-100, Campinas, Brazil.,Present address: Instituto Butantan & Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, Brazil
| | - Lucio H Freitas-Junior
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), 13083-100, Campinas, Brazil.,Present address: Instituto Butantan & Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, Brazil
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology ScreeningPort, 22525, Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Molecular Biology and Applied Ecology ScreeningPort, 22525, Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology ScreeningPort, 22525, Hamburg, Germany
| | - Chiara Borsari
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Maria Paola Costi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| |
Collapse
|
97
|
Gulin JEN, Bisio M, Rocco DM, Altcheh J, Solana ME, García-Bournissen F. Molecular and biological characterization of a highly pathogenic Trypanosoma cruzi strain isolated from a patient with congenital infection. Exp Parasitol 2018; 186:50-58. [PMID: 29448038 DOI: 10.1016/j.exppara.2018.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/23/2017] [Accepted: 02/11/2018] [Indexed: 11/16/2022]
Abstract
Although many Trypanosoma cruzi (T. cruzi) strains isolated from a wide range of hosts have been characterized, there is a lack of information about biological features from vertically transmitted strains. We describe the molecular and biological characteristics of the T. cruzi VD strain isolated from a congenital Chagas disease patient. The VD strain was typified as DTU TcVI; in vitro sensitivity to nifurtimox (NFX) and beznidazole (BZ) were 2.88 μM and 6.19 μM respectively, while inhibitory concentrations for intracellular amastigotes were 0.24 μM for BZ, and 0.66 μM for NFX. Biological behavior of VD strain was studied in a mouse model of acute infection, resulting in high levels of parasitemia and mortality with a rapid clearence of bloodstream trypomastigotes when treated with BZ or NFX, preventing mortality and reducing parasitic load and intensity of inflammatory infiltrate in skeletal and cardiac muscle. Treatment-induced parasitological cure, evaluated after immunossupression were 41% and 35% for BZ and NFX treatment respectively, suggesting a partial response to these drugs in elimination of parasite burden. This exhaustive characterization of this T. cruzi strain provides the basis for inclusion of this strain in a panel of reference strains for drug screening and adds a new valuable tool for the study of experimental T. cruzi infection.
Collapse
Affiliation(s)
- Julián Ernesto Nicolás Gulin
- Servicio de Parasitología y enfermedad de Chagas - Hospital General de Niños "Dr. Ricardo Gutiérrez", Gallo 1330, CP: 1425, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina.
| | - Margarita Bisio
- Servicio de Parasitología y enfermedad de Chagas - Hospital General de Niños "Dr. Ricardo Gutiérrez", Gallo 1330, CP: 1425, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Daniela Marisa Rocco
- Servicio de Parasitología y enfermedad de Chagas - Hospital General de Niños "Dr. Ricardo Gutiérrez", Gallo 1330, CP: 1425, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Jaime Altcheh
- Servicio de Parasitología y enfermedad de Chagas - Hospital General de Niños "Dr. Ricardo Gutiérrez", Gallo 1330, CP: 1425, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - María Elisa Solana
- Facultad de Medicina, Instituto de Investigaciones en Microbiología y Parasitología Médicas (IMPaM), UBA-CONICET, Universidad de Buenos Aires, Paraguay 2155, CP: 1121, Ciudad Autónoma de Buenos Aires, Argentina; Departamento de Cs. Básicas, Universidad Nacional de Luján, Ruta 5 y Avenida Constitución - (6700) Luján, Buenos Aires, Argentina
| | - Facundo García-Bournissen
- Servicio de Parasitología y enfermedad de Chagas - Hospital General de Niños "Dr. Ricardo Gutiérrez", Gallo 1330, CP: 1425, Ciudad Autónoma de Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| |
Collapse
|
98
|
Linciano P, Moraes CB, Alcantara LM, Franco CH, Pascoalino B, Freitas-Junior LH, Macedo S, Santarem N, Cordeiro-da-Silva A, Gul S, Witt G, Kuzikov M, Ellinger B, Ferrari S, Luciani R, Quotadamo A, Costantino L, Costi MP. Aryl thiosemicarbazones for the treatment of trypanosomatidic infections. Eur J Med Chem 2018; 146:423-434. [PMID: 29407968 DOI: 10.1016/j.ejmech.2018.01.043] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 01/12/2018] [Accepted: 01/14/2018] [Indexed: 01/07/2023]
Abstract
Basing on a library of thiadiazole derivatives showing anti-trypanosomatidic activity, we have considered the thiadiazoles opened forms and reaction intermediates, thiosemicarbazones, as compounds of interest for phenotypic screening against Trypanosoma brucei (Tb), intracellular amastigote form of Leishmania infantum (Li) and Trypanosoma cruzi (Tc). Similar compounds have already shown interesting activity against the same organisms. The compounds were particularly effective against T. brucei and T. cruzi. Among the 28 synthesized compounds, the best one was (E)-2-(4-((3.4-dichlorobenzyl)oxy)benzylidene) hydrazinecarbothioamide (A14) yielding a comparable anti-parasitic activity against the three parasitic species (TbEC50 = 2.31 μM, LiEC50 = 6.14 μM, TcEC50 = 1.31 μM) and a Selectivity Index higher than 10 with respect to human macrophages, therefore showing a pan-anti-trypanosomatidic activity. (E)-2-((3'.4'-dimethoxy-[1.1'-biphenyl]-3-yl)methyle ne) hydrazinecarbothioamide (A12) and (E)-2-(4-((3.4-dichlorobenzyl)oxy)benzylidene)hydrazine carbothioamide (A14) were able to potentiate the anti-parasitic activity of methotrexate (MTX) when evaluated in combination against T. brucei, yielding a 6-fold and 4-fold respectively Dose Reduction Index for MTX. The toxicity profile against four human cell lines and a panel of in vitro early-toxicity assays (comprising hERG, Aurora B, five cytochrome P450 isoforms and mitochondrial toxicity) demonstrated the low toxicity for the thosemicarbazones class in comparison with known drugs. The results confirmed thiosemicarbazones as a suitable chemical scaffold with potential for the development of properly decorated new anti-parasitic drugs.
Collapse
Affiliation(s)
- Pasquale Linciano
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Carolina B Moraes
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas, SP, Brazil
| | - Laura M Alcantara
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas, SP, Brazil
| | - Caio H Franco
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas, SP, Brazil
| | - Bruno Pascoalino
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas, SP, Brazil
| | - Lucio H Freitas-Junior
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas, SP, Brazil
| | - Sara Macedo
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal; Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Nuno Santarem
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal; Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Anabela Cordeiro-da-Silva
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal; Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal; Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, Hamburg, Germany
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, Hamburg, Germany
| | - Stefania Ferrari
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Rosaria Luciani
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Antonio Quotadamo
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Luca Costantino
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Maria Paola Costi
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy.
| |
Collapse
|
99
|
Inhibitors of Trypanosoma cruzi Sir2 related protein 1 as potential drugs against Chagas disease. PLoS Negl Trop Dis 2018; 12:e0006180. [PMID: 29357372 PMCID: PMC5794198 DOI: 10.1371/journal.pntd.0006180] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 02/01/2018] [Accepted: 12/19/2017] [Indexed: 11/26/2022] Open
Abstract
Chagas disease remains one of the most neglected diseases in the world despite being the most important parasitic disease in Latin America. The characteristic chronic manifestation of chagasic cardiomyopathy is the region’s leading cause of heart-related illness, causing significant mortality and morbidity. Due to the limited available therapeutic options, new drugs are urgently needed to control the disease. Sirtuins, also called Silent information regulator 2 (Sir2) proteins have long been suggested as interesting targets to treat different diseases, including parasitic infections. Recent studies on Trypanosoma cruzi sirtuins have hinted at the possibility to exploit these enzymes as a possible drug targets. In the present work, the T. cruzi Sir2 related protein 1 (TcSir2rp1) is genetically validated as a drug target and biochemically characterized for its NAD+-dependent deacetylase activity and its inhibition by the classic sirtuin inhibitor nicotinamide, as well as by bisnaphthalimidopropyl (BNIP) derivatives, a class of parasite sirtuin inhibitors. BNIPs ability to inhibit TcSir2rp1, and anti-parasitic activity against T. cruzi amastigotes in vitro were investigated. The compound BNIP Spermidine (BNIPSpd) (9), was found to be the most potent inhibitor of TcSir2rp1. Moreover, this compound showed altered trypanocidal activity against TcSir2rp1 overexpressing epimastigotes and anti-parasitic activity similar to the reference drug benznidazole against the medically important amastigotes, while having the highest selectivity index amongst the compounds tested. Unfortunately, BNIPSpd failed to treat a mouse model of Chagas disease, possibly due to its pharmacokinetic profile. Medicinal chemistry modifications of the compound, as well as alternative formulations may improve activity and pharmacokinetics in the future. Additionally, an initial TcSIR2rp1 model in complex with p53 peptide substrate was obtained from low resolution X-ray data (3.5 Å) to gain insight into the potential specificity of the interaction with the BNIP compounds. In conclusion, the search for TcSir2rp1 specific inhibitors may represent a valuable strategy for drug discovery against T. cruzi. Trypanosoma cruzi is a protozoan parasite belonging to the Kinetoplastida class responsible for Chagas disease, a neglected tropical illness that affects an estimated 6 to 8 million people in Latin America and some Southern regions of the USA, with another 25 million at risk of acquiring the disease and a death toll of 12,000 every year. Commonly transmitted from the feces of the kissing bug, the disease is characterized by a nearly asymptomatic acute phase but a problematic chronic phase in which 20–30% of individuals develop serious cardiac and/or intestinal problems. The therapies currently in use were introduced more than forty years ago, and there are important concerns about adverse effects and lower effectiveness with disease progression. There is, therefore, an urgent need to find better alternatives. In this study, we evaluate the potential of a Trypanosoma cruzi sirtuin protein as a novel drug target and its inhibition by novel members of a known class of sirtuin compound inhibitors.
Collapse
|
100
|
Franco J, Scarone L, Comini MA. Drugs and Drug Resistance in African and American Trypanosomiasis. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2018. [DOI: 10.1016/bs.armc.2018.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|