1
|
de Oliveira R, Cruz LR, Dessoy MA, Koovits PJ, dos Santos DA, de Oliveira LFN, Ferreira RA, Mollo MC, Lee E, Duarte SM, Krogh R, Ferreira LLG, Chelucci RC, Dichiara M, Simpson QJ, Feltrin C, da Silva AC, dos Santos BM, Broering MF, Pollastri MP, Ferrins L, Moraes CB, Andricopulo AD, Kratz JM, Sjö P, Mowbray CE, Dias LC. Discovery and Early Optimization of 1 H-Indole-2-carboxamides with Anti- Trypanosoma cruzi Activity. J Med Chem 2025; 68:7313-7340. [PMID: 40163677 PMCID: PMC11998015 DOI: 10.1021/acs.jmedchem.4c02942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/08/2025] [Accepted: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Chagas disease (CD), caused by the flagellate protozoan Trypanosoma cruzi, is a neglected tropical disease endemic in 21 countries. The only two antiparasitic drugs approved for its treatment, benznidazole and nifurtimox, have significant drawbacks. We present herein the optimization of a series of substituted indoles that were identified through phenotypic screening against T. cruzi. Early lead compounds with balanced potency and physicochemical properties were advanced to animal studies but showed limited plasma exposure. Medicinal chemistry strategies were used to improve metabolic stability and solubility, but unfortunately, this effort failed to yield compounds with improvements in both exposure and potency. Still, the best compound was progressed for a proof-of-concept efficacy study using acute and chronic mice models of Chagas disease. Despite showing antiparasitic activity in these in vivo studies, the optimization work with this series was stopped due to unfavorable drug metabolism and pharmacokinetic (DMPK) properties and a deprioritized mechanism of action (CYP51 inhibition).
Collapse
Affiliation(s)
- Ramon
G. de Oliveira
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | - Luiza R. Cruz
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
- Drugs
for Neglected Diseases initiative, Rio de Janeiro 20010-020, Brazil
| | - Marco A. Dessoy
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | - Paul J. Koovits
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | | | | | - Rafael A. Ferreira
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | - María C. Mollo
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | - Eun Lee
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| | - Simone M. Duarte
- São
Carlos Institute of Physics, University
of São Paulo, São
Carlos 13563-120, Brazil
| | - Renata Krogh
- São
Carlos Institute of Physics, University
of São Paulo, São
Carlos 13563-120, Brazil
| | | | - Rafael C. Chelucci
- São
Carlos Institute of Physics, University
of São Paulo, São
Carlos 13563-120, Brazil
| | - Maria Dichiara
- Chemistry and Chemical Biology and Pharmaceutical
Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Quillon J. Simpson
- Chemistry and Chemical Biology and Pharmaceutical
Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Clarissa Feltrin
- Institute of Biomedical Sciences and School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Adriana C. da Silva
- Institute of Biomedical Sciences and School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Benedito M. dos Santos
- Institute of Biomedical Sciences and School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Milena F. Broering
- Institute of Biomedical Sciences and School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Michael P. Pollastri
- Chemistry and Chemical Biology and Pharmaceutical
Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Lori Ferrins
- Chemistry and Chemical Biology and Pharmaceutical
Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Carolina B. Moraes
- Institute of Biomedical Sciences and School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Jadel M. Kratz
- Drugs
for Neglected Diseases initiative, Rio de Janeiro 20010-020, Brazil
| | - Peter Sjö
- Drugs
for Neglected Diseases initiative, Geneva 1202, Switzerland
| | | | - Luiz C. Dias
- Institute
of Chemistry, State University of Campinas, Campinas 13083-862, Brazil
| |
Collapse
|
2
|
de Souza GA, Chaves LDS, Velez ASMM, Lacerda JLF, Pitasse-Santos P, dos Santos JCC, Chaves OA, Serpa C, Valente RDC, da Fonseca LM, da Costa Santos MAR, dos Reis JS, Santos CADN, Mendonça-Previato L, Previato JO, Freire-de-Lima CG, Decoté-Ricardo D, Freire-de-Lima L, de Lima MEF. Design and Synthesis of Bis-Chalcones as Curcumin Simplified Analogs and Assessment of Their Antiproliferative Activities Against Human Lung Cancer Cells and Trypanosoma cruzi Amastigotes. Pharmaceuticals (Basel) 2025; 18:456. [PMID: 40283893 PMCID: PMC12030033 DOI: 10.3390/ph18040456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Anticancer therapies represent the primary treatment option for a significant number of cancer patients globally; however, many of these treatments are associated with severe side effects as they target molecular structures present in both cancerous and healthy cells. In a similar context, the treatment of Chagas disease, a neglected tropical illness, is hindered by the high toxicity of the currently available drugs. Researchers are increasingly focusing on the development of safer and more selective alternatives, with natural compounds being studied as potential starting points for the creation of more effective drug candidates with a favorable therapeutic index. Objectives: The aim of this study was to design simplified curcumin-derived structures that preserved or enhanced their therapeutic activity against human lung cancer cell lines and T. cruzi, while also improving bioavailability and minimizing toxicity. Methods: In this study, curcumin and two natural curcuminoids inspired the synthesis of a chalcone and a set of bis-chalcones, compound classes known for their enhanced stability compared with their natural parent derivatives. The synthetic strategy used was the acid-catalyzed aldol condensation reaction. The stability profiles, IC50 values against A549 and H460 tumor cell lines, and trypanocidal activity against T. cruzi amastigotes of these derivatives were assessed. Results: The synthesized derivatives exhibited improved stability compared with the parent compounds, along with lower IC50 values in both A549 and H460 tumor cell lines. Additionally, one of the new analogs showed promising trypanocidal activity against T. cruzi amastigotes. Conclusions: This study provides a potential pathway toward the development of more effective and less toxic treatments for both cancer and Chagas disease. The simplified curcumin derivatives represent a promising foundation for designing new therapeutic agents with improved bioavailability and efficacy.
Collapse
Affiliation(s)
- Gabriela Alves de Souza
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil; (G.A.d.S.); (A.S.M.M.V.); (J.L.F.L.); (J.C.C.d.S.)
| | - Lorrane de Souza Chaves
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Afonso Santine M. M. Velez
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil; (G.A.d.S.); (A.S.M.M.V.); (J.L.F.L.); (J.C.C.d.S.)
| | - Jorge Lucas F. Lacerda
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil; (G.A.d.S.); (A.S.M.M.V.); (J.L.F.L.); (J.C.C.d.S.)
| | - Paulo Pitasse-Santos
- Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester LE1 7HB, UK;
- School of Chemistry, University of Leicester, Leicester LE1 7RH, UK
| | - Jayane Clys Conceição dos Santos
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil; (G.A.d.S.); (A.S.M.M.V.); (J.L.F.L.); (J.C.C.d.S.)
| | - Otávio Augusto Chaves
- Department of Chemistry, Coimbra Chemistry Centre-Institute of Molecular Science, University of Coimbra, 3004-535 Coimbra, Portugal; (O.A.C.); (C.S.)
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias, Oswaldo Cruz Institute, Rio de Janeiro 21040-361, RJ, Brazil
| | - Carlos Serpa
- Department of Chemistry, Coimbra Chemistry Centre-Institute of Molecular Science, University of Coimbra, 3004-535 Coimbra, Portugal; (O.A.C.); (C.S.)
| | - Raphael do Carmo Valente
- Campus Duque de Caxias Professor Geraldo Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias 25.240-005, RJ, Brazil;
| | - Leonardo Marques da Fonseca
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
- Curso de Medicina, Universidade Castelo Branco, Rio de Janeiro 21.710-255, RJ, Brazil
| | - Marcos André Rodrigues da Costa Santos
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Jhenifer Santos dos Reis
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Carlos Antônio do Nascimento Santos
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Lucia Mendonça-Previato
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Jose Osvaldo Previato
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Celio Geraldo Freire-de-Lima
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
| | - Debora Decoté-Ricardo
- Departamento de Microbiologia e Imunologia Veterinária, Instituto de Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil;
| | - Leonardo Freire-de-Lima
- Programa de Pós-Graduação em Ciências Biológicas (Biofísica), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (L.d.S.C.); (L.M.d.F.); (M.A.R.d.C.S.); (J.S.d.R.); (C.A.d.N.S.); (L.M.-P.); (J.O.P.); (C.G.F.-d.-L.)
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Programa de Pós-Graduação em Ciências Morfológicas, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Marco Edilson Freire de Lima
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Rural do Rio de Janeiro, Seropédica 23.897-000, RJ, Brazil; (G.A.d.S.); (A.S.M.M.V.); (J.L.F.L.); (J.C.C.d.S.)
| |
Collapse
|
3
|
Camargo PG, da Silva RB, Zuma AA, Garden SJ, Albuquerque MG, Rodrigues CR, da Silva Lima CH. In silico evaluation of N-aryl-1,10-phenanthroline-2-amines as potential inhibitors of T. cruzi GP63 zinc-metalloprotease by docking and molecular dynamics simulations. Sci Rep 2025; 15:6036. [PMID: 39971997 PMCID: PMC11839977 DOI: 10.1038/s41598-025-90088-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/10/2025] [Indexed: 02/21/2025] Open
Abstract
Based on the in vitro trypanocidal efficacy of previously synthesized N-aryl-1,10-phenanthroline-2-amines (Phen1-20) (aryl = R-phenyl, 1- or 2-naphthyl), we explored the potential interactions of these derivatives as ligands of our comparative model of T. cruzi GP63 (TcGP63). This surface metalloprotease plays a crucial role in parasite adhesion to host cells and aids in cell invasion during T. cruzi infection in Chagas disease. Ligand-protein consensus docking simulations using four GOLD scoring functions revealed that N-(R-phenyl) derivatives (R = CH3, OCH3, CF3, CN, NO2, F, Cl, and Br) presented poses with higher fitness scores than the N-naphthyl ones, with the six para-substituted derivatives (Phen4, p-CH3; Phen7, p-OCH3; Phen10, p-CN; Phen14, p-F; Phen17, p-Cl; and Phen18, p-Br) being more favorable than the ortho or meta ones. Subsequent aqueous molecular dynamics simulation (GROMACS package, CHARMM36 force field, and TIP3P water model) of the ligand-protein complexes for these six top-ranking compounds showed persistent interactions within the TcGP63 active site, primarily through coordination with Zn(II)-cofactor, and H-bonding with catalytic Glu221 and zinc-binding His224. RMSD and RMSF analyses confirmed the stability of these interactions, particularly for compounds with electron-withdrawing groups by inductive effect as R-substituents, such as p-OCH3 (Phen7) and p-CN (Phen10). Binding free energy calculations by the linear interaction energy (LIE) approach corroborated the favorable interactions observed in simulations, highlighting Phen7 and Phen10 as the most promising candidates. This study underscores the potential of N-phenyl-1,10-phenanthroline-2-amines as putative inhibitors targeting the T. cruzi GP63 enzyme.
Collapse
Affiliation(s)
- Priscila Goes Camargo
- Laboratório de Modelagem Molecular e QSAR (ModMolQSAR), Departamento de Fármacos e Medicamentos, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ramon Borges da Silva
- Laboratório Nacional de Biociências Brasileiras, Centro Brasileiro de Pesquisa em Energia e Materiais, Campinas, SP, Brazil
| | - Aline Araujo Zuma
- Laboratório de Ultraestrutura Celular Hertha Meyer, Centro de Pesquisas em Medicina de Precisão, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Simon J Garden
- Departamento de Química Orgânica, Programa de Pós-Graduação em Química (PGQu), Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Magaly Girão Albuquerque
- Departamento de Química Orgânica, Programa de Pós-Graduação em Química (PGQu), Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carlos Rangel Rodrigues
- Laboratório de Modelagem Molecular e QSAR (ModMolQSAR), Departamento de Fármacos e Medicamentos, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Camilo Henrique da Silva Lima
- Departamento de Química Orgânica, Programa de Pós-Graduação em Química (PGQu), Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
4
|
Gonçalves ACR, Libardi SH, Borges JC, Oliveira RJ, Gotzmann C, Blacque O, de Albuquerque S, Lopes CD, Alberto R, Maia PIS. Rhenium(I) and technetium(I) complexes with megazol derivatives: towards the development of a theranostic platform for Chagas disease. Dalton Trans 2024; 53:19153-19165. [PMID: 39533923 DOI: 10.1039/d4dt02714k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The diagnosis and treatment of Chagas disease (CD) in the chronic phase remains a challenge. With that in mind, a potential theranostic device based on the trypanocidal agent known as megazol and the fac-M(CO)3+ (M = Re or 99mTc) fragment is proposed in the present work. The peripheral structure of megazol (LH,H) was modified to obtain the compounds LR1,R2 (R1 = H, R2 = Me and R1 = R2 = Me), which were used in the syntheses of complexes of composition [ReBr(CO)3LR1,R2]. These compounds were studied by elemental analysis, FTIR, UV-vis, NMR (1H and 13C), HR-ESI-MS, HPLC/UPLC and single-crystal XRD. The trypanocidal activity of the rhenium complexes was evaluated in vitro against the intracellular form of Trypanosoma cruzi. With exception of the [ReBr(CO)3LMe,Me] complex, all compounds are more active than the standard drug, benznidazole (Bzn), while [ReBr(CO)3LH,H] also exhibited a much higher selectivity index. In addition, the interaction of megazol and its ReI complex was evaluated with the T. cruzi Old Yellow Enzyme (TcOYE) by both experimental and computational methods. The data showed that megazol as well as its metal complex exhibited a higher affinity for TcOYE compared to Bzn. Finally, the labelling of megazol with 99mTc was successfully carried out. However, the results indicated that the Re complexes used as standards were not homologous with the 99mTc complexes. Despite this discrepancy, this research suggests that the investigation into Re and 99mTc complexes with megazol could lead to the development of a theranostic device for CD in a near future.
Collapse
Affiliation(s)
- Ana C R Gonçalves
- Bioactive Compounds Development Research Group, Federal University of Triângulo Mineiro, Av. Dr. Ran-dolfo Borges 1400, Uberaba 38025-440, MG, Brazil
| | - Silvia H Libardi
- São Carlos Institute of Chemistry, University of São Paulo, Av. Trabalhador São Carlense, 400, São Carlos 13566-590, SP, Brazil
| | - Júlio C Borges
- São Carlos Institute of Chemistry, University of São Paulo, Av. Trabalhador São Carlense, 400, São Carlos 13566-590, SP, Brazil
| | - Ronaldo J Oliveira
- Bioactive Compounds Development Research Group, Federal University of Triângulo Mineiro, Av. Dr. Ran-dolfo Borges 1400, Uberaba 38025-440, MG, Brazil
| | - Carla Gotzmann
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Olivier Blacque
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Sergio de Albuquerque
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto - FCFRP-USP, University of São Paulo, Av. do Café s/n, 14040-903, Ribeirão Preto, SP, Brazil
| | - Carla D Lopes
- Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirão Preto - FCFRP-USP, University of São Paulo, Av. do Café s/n, 14040-903, Ribeirão Preto, SP, Brazil
- Ribeirão Preto's Estácio University Center, Abraão Issa Halach Street, 980, 14096-160, Ribeirão Preto, SP, Brazil
| | - Roger Alberto
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Pedro I S Maia
- Bioactive Compounds Development Research Group, Federal University of Triângulo Mineiro, Av. Dr. Ran-dolfo Borges 1400, Uberaba 38025-440, MG, Brazil
| |
Collapse
|
5
|
Nwoke EA, Lowe S, Aldabbagh F, Kalesh K, Kadri H. Nucleoside Analogues for Chagas Disease and Leishmaniasis Therapy: Current Status and Future Perspectives. Molecules 2024; 29:5234. [PMID: 39598623 PMCID: PMC11596272 DOI: 10.3390/molecules29225234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024] Open
Abstract
Chagas disease and leishmaniasis are two neglected tropical diseases that affect millions of people in low- and middle-income tropical countries. These diseases caused by protozoan parasites pose significant global health challenges, which have been exacerbated by the recent COVID-19 pandemic. There is an urgent need for novel therapeutics as current treatments are limited by toxicity and drug resistance. Nucleoside analogues, which have been extensively studied and successfully applied in antiviral and antitumor therapies, hold potential that has yet to be fully explored for treating these neglected diseases. In this review, we discuss the use of nucleoside analogues as promising therapeutic agents for Chagas disease and leishmaniasis. After briefly examining the pathology, progression, and current treatment options for these diseases, we provide a comprehensive analysis of the status of nucleoside analogues and explore their prospects. By outlining the current landscape and future directions, this review aims to guide research and development efforts towards more effective nucleoside-based treatments for Chagas disease and leishmaniasis.
Collapse
Affiliation(s)
- Emmanuel Awucha Nwoke
- Department of Pharmacy, School of Life Sciences, Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, London KT1 2EE, UK; (E.A.N.); (S.L.); (F.A.)
| | - Silvester Lowe
- Department of Pharmacy, School of Life Sciences, Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, London KT1 2EE, UK; (E.A.N.); (S.L.); (F.A.)
| | - Fawaz Aldabbagh
- Department of Pharmacy, School of Life Sciences, Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, London KT1 2EE, UK; (E.A.N.); (S.L.); (F.A.)
| | - Karunakaran Kalesh
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK;
| | - Hachemi Kadri
- Department of Pharmacy, School of Life Sciences, Pharmacy and Chemistry, Kingston University, Penrhyn Road, Kingston upon Thames, London KT1 2EE, UK; (E.A.N.); (S.L.); (F.A.)
| |
Collapse
|
6
|
Espinoza-Chávez RM, de Oliveira Rezende Júnior C, Laureano de Souza M, Consolin Chelucci R, Michelan-Duarte S, Krogh R, Gomes Ferreira LL, Valli M, Sena de Oliveira A, Andricopulo AD, Carlos Dias L. Structure-Metabolism Relationships of Benzimidazole Derivatives with anti-Trypanosoma cruzi Activity for Chagas Disease. ChemMedChem 2024; 19:e202400293. [PMID: 38924252 DOI: 10.1002/cmdc.202400293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
This study introduces further insights from the hit-to-lead optimization process involving a series of benzimidazole derivatives acting as inhibitors of the cruzain enzyme, which targets Trypanosoma cruzi, the causative parasite of Chagas disease. Here, we present the design, synthesis and biological evaluation of 30 new compounds as a third generation of benzimidazole analogues with trypanocidal activity, aiming to enhance our understanding of their pharmacokinetic profiles and establish a structure-metabolism relationships within the series. The design of these new analogues was guided by the analysis of previous pharmacokinetic results, considering identified metabolic sites and biotransformation studies. This optimization resulted in the discovery of two compounds (42 e and 49 b) exhibiting enhanced metabolic stability, anti-Trypanosoma cruzi activity compared to benznidazole (the reference drug for Chagas disease), as well as being non-cruzain inhibitors, and demonstrating a satisfactory in vitro pharmacokinetic profile. These findings unveil a new subclass of aminobenzimidazole and rigid compounds, which offer potential for further exploration in the quest for discovering novel classes of antichagasic compounds.
Collapse
Affiliation(s)
- Rocío Marisol Espinoza-Chávez
- Laboratory of Synthetic Organic Chemistry, Institute of Chemistry, State University of Campinas (Unicamp), Campinas-SP, 13084-971, Brazil
| | - Celso de Oliveira Rezende Júnior
- Laboratory of Synthetic Organic Chemistry, Institute of Chemistry, State University of Campinas (Unicamp), Campinas-SP, 13084-971, Brazil
- Institute of Chemistry, Federal University of Uberlândia (UFU), Uberlândia-MG, 38400-902, Brazil
| | - Mariana Laureano de Souza
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos (IFSC), University of Sao Paulo (USP), Sao Carlos-SP, 13563-120, Brazil
| | - Rafael Consolin Chelucci
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos (IFSC), University of Sao Paulo (USP), Sao Carlos-SP, 13563-120, Brazil
| | - Simone Michelan-Duarte
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos (IFSC), University of Sao Paulo (USP), Sao Carlos-SP, 13563-120, Brazil
| | - Renata Krogh
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos (IFSC), University of Sao Paulo (USP), Sao Carlos-SP, 13563-120, Brazil
| | - Leonardo Luiz Gomes Ferreira
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos (IFSC), University of Sao Paulo (USP), Sao Carlos-SP, 13563-120, Brazil
| | - Marilia Valli
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos (IFSC), University of Sao Paulo (USP), Sao Carlos-SP, 13563-120, Brazil
| | - Aldo Sena de Oliveira
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos (IFSC), University of Sao Paulo (USP), Sao Carlos-SP, 13563-120, Brazil
- Department of Exact Sciences and Education, Federal University of Santa Catarina (UFSC), Blumenau-SC, 89036-004, Brazil
| | - Adriano D Andricopulo
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of Sao Carlos (IFSC), University of Sao Paulo (USP), Sao Carlos-SP, 13563-120, Brazil
| | - Luiz Carlos Dias
- Laboratory of Synthetic Organic Chemistry, Institute of Chemistry, State University of Campinas (Unicamp), Campinas-SP, 13084-971, Brazil
| |
Collapse
|
7
|
Alonso VL, Escalante AM, Rodríguez Araya E, Frattini G, Tavernelli LE, Moreno DM, Furlan RLE, Serra E. 1,3,4-oxadiazoles as inhibitors of the atypical member of the BET family bromodomain factor 3 from Trypanosoma cruzi ( TcBDF3). Front Microbiol 2024; 15:1465672. [PMID: 39411427 PMCID: PMC11473290 DOI: 10.3389/fmicb.2024.1465672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, affects millions globally, with increasing urban cases outside of Latin America. Treatment is based on two compounds, namely, benznidazole (BZ) and nifurtimox, but chronic cases pose several challenges. Targeting lysine acetylation, particularly bromodomain-containing proteins, shows promise as a novel antiparasitic target. Our research focuses on TcBDF3, a cytoplasmic protein, which is crucial for parasite differentiation that recognizes acetylated alpha-tubulin. In our previous study, A1B4 was identified as a high-affinity binder of TcBDF3, showing significant trypanocidal activity with low host toxicity in vitro. In this report, the binding of TcBDF3 to A1B4 was validated using differential scanning fluorescence, fluorescence polarization, and molecular modeling, confirming its specific interaction. Additionally, two new 1,3,4-oxadiazoles derived from A1B4 were identified, which exhibited improved trypanocide activity and cytotoxicity profiles. Furthermore, TcBDF3 was classified for the first time as an atypical divergent member of the bromodomain extraterminal family found in protists and plants. These results make TcBDF3 a unique target due to its localization and known functions not shared with higher eukaryotes, which holds promise for Chagas disease treatment.
Collapse
Affiliation(s)
- Victoria L. Alonso
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, Rosario, Argentina
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Andrea M. Escalante
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Elvio Rodríguez Araya
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, Rosario, Argentina
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Gianfranco Frattini
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
- Instituto de Química Rosario, CONICET-UNR, Rosario, Argentina
| | - Luis E. Tavernelli
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, Rosario, Argentina
| | - Diego M. Moreno
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
- Instituto de Química Rosario, CONICET-UNR, Rosario, Argentina
| | - Ricardo L. E. Furlan
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Esteban Serra
- Instituto de Biología Molecular y Celular de Rosario, CONICET-UNR, Rosario, Argentina
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|
8
|
Jandl B, Zheng R, Muttenthaler M, Baell J. Novel Diacyl-hydrazide Compounds as Potential Therapeutics for Visceral Leishmaniasis. ACS OMEGA 2024; 9:37170-37182. [PMID: 39246504 PMCID: PMC11375810 DOI: 10.1021/acsomega.4c04517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 07/07/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024]
Abstract
Visceral leishmaniasis is a neglected tropical disease with the highest mortality among different forms of leishmaniasis manifestation in humans. The disease is caused by the parasitic protists Leishmania donovani and Leishmania infantum, and treatments remain unsuitable due to high costs, complicated administration, lack of efficacy, variable patient susceptibility, toxic side effects, and rising parasitic resistance. Herein, we report a structure-activity relationship (SAR) exploration of the diacyl-hydrazide scaffold identified to have antiparasitic activity from a high-throughput screen against L. donovani, Trypanosoma cruzi, and Trypanosoma brucei. This SAR study revealed new structural insights into this scaffold related to bioactivity resulting in a new series of lead compounds with nanomolar activity against L. donovani and no toxicity against human THP-1 macrophages. These optimized diacyl-hydrazide compounds set the stage for future drug development and hold promise for a new treatment avenue for visceral leishmaniasis.
Collapse
Affiliation(s)
- Bernhard Jandl
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Vienna Doctoral School in Chemistry, University of Vienna, 1090 Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, 4072 Brisbane, Queensland, Australia
| | - Rebecca Zheng
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
| | - Markus Muttenthaler
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, 4072 Brisbane, Queensland, Australia
| | - Jonathan Baell
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Victoria, Australia
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
- Australian Translational Medicinal Chemistry Facility, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
9
|
Tristão DC, Barbosa H, de Castro Levatti EV, Andrade BA, Romanelli MM, Antar GM, Tempone AG, Lago JHG. Selective Activity Against Amastigote Forms of Trypanosoma cruzi and Leishmania infantum of Diastereomeric Dicentrine N-oxides. Chem Biodivers 2024; 21:e202401247. [PMID: 38896778 DOI: 10.1002/cbdv.202401247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 06/21/2024]
Abstract
As part of our continuous research for the discovery of bioactive compounds against Trypanosoma cruzi and Leishmania infantum, the alkaloid (6aS)-dicentrine (1) was oxidized to afford (6aS,6S)- (2) and (6aS,6R)- (3) dicentrine-N-oxides. Evaluation of the cytotoxicity against NCTC cells indicated that 2 and 3 are non-toxic (CC50>200 μM) whereas 1 demonstrated CC50 of 52.0 μM. Concerning T. cruzi activity against amastigotes, derivatives 2 and 3 exhibited EC50 values of 9.9 μM (SI>20.2) and 27.5 μM (SI>7.3), respectively, but 1 is inactive (EC50>100 μM). Otherwise, when tested against L. infantum amastigotes, 1 and 3 exhibited EC50 values of 10.3 μM (SI=5.0) and 12.7 μM (SI>15.7), respectively, being 2 inactive (EC50>100 μM). Comparing the effects of positive controls benznidazol (EC50=6.5 μM and SI>30.7) and miltefosine (EC50=10.2 μM and SI=15.2), it was observed a selective antiparasitic activity to diastereomers 2 and 3 against T. cruzi and L. infantum. Considering stereochemical aspects, it was suggested that the configuration of the new stereocenter formed after oxidation of 1 played an important role in the bioactivity against amastigotes of both tested parasites.
Collapse
Affiliation(s)
- Daniela C Tristão
- Center for Natural and Human Sciences, Federal University of ABC, 09210-580, Santo Andre, SP, Brazil
| | - Henrique Barbosa
- Center for Natural and Human Sciences, Federal University of ABC, 09210-580, Santo Andre, SP, Brazil
| | | | | | | | - Guilherme M Antar
- Department of Agricultural and Biological Sciences, Federal University of Espirito Santo, 29932-540, São Matheus, ES, Brazil
| | | | - João Henrique G Lago
- Center for Natural and Human Sciences, Federal University of ABC, 09210-580, Santo Andre, SP, Brazil
| |
Collapse
|
10
|
Moncada-Basualto M, Saavedra-Olavarría J, Rivero-Jerez PS, Rojas C, Maya JD, Liempi A, Zúñiga-Bustos M, Olea-Azar C, Lapier M, Pérez EG, Pozo-Martínez J. Assessment of the Activity of Nitroisoxazole Derivatives against Trypanosoma cruzi. Molecules 2024; 29:2762. [PMID: 38930828 PMCID: PMC11207111 DOI: 10.3390/molecules29122762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
The development of new compounds to treat Chagas disease is imperative due to the adverse effects of current drugs and their low efficacy in the chronic phase. This study aims to investigate nitroisoxazole derivatives that produce oxidative stress while modifying the compounds' lipophilicity, affecting their ability to fight trypanosomes. The results indicate that these compounds are more effective against the epimastigote form of T. cruzi, with a 52 ± 4% trypanocidal effect for compound 9. However, they are less effective against the trypomastigote form, with a 15 ± 3% trypanocidal effect. Additionally, compound 11 interacts with a higher number of amino acid residues within the active site of the enzyme cruzipain. Furthermore, it was also found that the presence of a nitro group allows for the generation of free radicals; likewise, the large size of the compound enables increased interaction with aminoacidic residues in the active site of cruzipain, contributing to trypanocidal activity. This activity depends on the size and lipophilicity of the compounds. The study recommends exploring new compounds based on the nitroisoxazole skeleton, with larger substituents and lipophilicity to enhance their trypanocidal activity.
Collapse
Affiliation(s)
- Mauricio Moncada-Basualto
- Instituto Universitario de Investigación y Desarrollo Tecnológico, Universidad Tecnológica Metropolitana, San Joaquín 8940577, Chile; (M.M.-B.); (C.R.); (M.Z.-B.)
| | - Jorge Saavedra-Olavarría
- Department of Organic Chemistry, Faculty of Chemistry and Pharmacy, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, San Joaquin 7820436, Chile; (J.S.-O.); (P.S.R.-J.)
| | - Paula S. Rivero-Jerez
- Department of Organic Chemistry, Faculty of Chemistry and Pharmacy, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, San Joaquin 7820436, Chile; (J.S.-O.); (P.S.R.-J.)
| | - Cristian Rojas
- Instituto Universitario de Investigación y Desarrollo Tecnológico, Universidad Tecnológica Metropolitana, San Joaquín 8940577, Chile; (M.M.-B.); (C.R.); (M.Z.-B.)
- Laboratory of Free Radicals and Antioxidants, Faculty of Chemical and Pharmaceutical Science, Universidad de Chile, Olivos 1007, Independencia 8380544, Chile;
| | - Juan D. Maya
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia 8380453, Chile;
| | - Ana Liempi
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia 8380453, Chile;
| | - Matías Zúñiga-Bustos
- Instituto Universitario de Investigación y Desarrollo Tecnológico, Universidad Tecnológica Metropolitana, San Joaquín 8940577, Chile; (M.M.-B.); (C.R.); (M.Z.-B.)
| | - Claudio Olea-Azar
- Laboratory of Free Radicals and Antioxidants, Faculty of Chemical and Pharmaceutical Science, Universidad de Chile, Olivos 1007, Independencia 8380544, Chile;
| | - Michel Lapier
- Centro de Investigación, Desarrollo e Innovación de Productos Bioactivos (CinBio), Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Av. Gran Bretaña 1093, Valparaiso 2360102, Chile;
| | - Edwin G. Pérez
- Department of Organic Chemistry, Faculty of Chemistry and Pharmacy, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, San Joaquin 7820436, Chile; (J.S.-O.); (P.S.R.-J.)
| | - Josué Pozo-Martínez
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Independencia 8380453, Chile;
- Laboratorio de Química—Médica, Facultad de Ciencia y Tecnología, Universidad del Azuay, Av. 24 de Mayo 777, Cuenca 010204, Ecuador
| |
Collapse
|
11
|
Murta SMF, Lemos Santana PA, Jacques Dit Lapierre TJW, Penteado AB, El Hajje M, Navarro Vinha TC, Liarte DB, de Souza ML, Goulart Trossini GH, de Oliveira Rezende Júnior C, de Oliveira RB, Ferreira RS. New drug discovery strategies for the treatment of benznidazole-resistance in Trypanosoma cruzi, the causative agent of Chagas disease. Expert Opin Drug Discov 2024; 19:741-753. [PMID: 38715393 DOI: 10.1080/17460441.2024.2349155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/25/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Benznidazole, the drug of choice for treating Chagas Disease (CD), has significant limitations, such as poor cure efficacy, mainly in the chronic phase of CD, association with side effects, and parasite resistance. Understanding parasite resistance to benznidazole is crucial for developing new drugs to treat CD. AREAS COVERED Here, the authors review the current understanding of the molecular basis of benznidazole resistance. Furthermore, they discuss the state-of-the-art methods and critical outcomes employed to evaluate the efficacy of potential drugs against T. cruzi, aiming to select better compounds likely to succeed in the clinic. Finally, the authors describe the different strategies employed to overcome resistance to benznidazole and find effective new treatments for CD. EXPERT OPINION Resistance to benznidazole is a complex phenomenon that occurs naturally among T. cruzi strains. The combination of compounds that inhibit different metabolic pathways of the parasite is an important strategy for developing a new chemotherapeutic protocol.
Collapse
Affiliation(s)
- Silvane Maria Fonseca Murta
- Grupo de Genômica Funcional de Parasitos - Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Pedro Augusto Lemos Santana
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - André Berndt Penteado
- Departamento de Farmacia, Faculdade de Ciencias Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Marissa El Hajje
- Departamento de Farmacia, Faculdade de Ciencias Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Mariana Laureano de Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | | | - Renata Barbosa de Oliveira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Rafaela Salgado Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
12
|
de M Gonçalves M, Totini CH, De Castro EV, Albuquerque V, Antar GM, Tempone AG, Lago JHG. Lignans Isolated from Piper truncatum Act as New Potent Antitrypanosomal Compounds. Chem Biodivers 2024; 21:e202400547. [PMID: 38507773 DOI: 10.1002/cbdv.202400547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 03/22/2024]
Abstract
The hexane extract from twigs of Piper truncatum Vell (Piperaceae) displayed activity against Trypanosoma cruzi and was subjected to chromatographic steps to afford six dibenzylbutyrolactolic lignans, being four knowns: cubebin (1), (-)-9α-O-methylcubebin (2), (+)-9β-O-methylcubebinin (3) and 3,4-dimethoxy-3,4-demethylenedioxycubebin (4) as well as two new, named truncatin A (5) and B (6). Initially, in vitro activity against trypomastigotes was evaluated and compounds 1, 4 and 6 exhibited EC50 values of 41.6, 21.0 and 39.6 μM, respectively. However, when tested against amastigotes, the relevant clinical form in the chronic phase of Chagas disease, compounds 1-6 displayed activities with EC50 values ranging from 1.6 to 13.7 μM. In addition, the mammalian cytotoxicity of compounds 1-6 was evaluated against murine fibroblasts (NCTC). Compounds 2, 3 and 4 exhibited reduced toxicity against NCTC cells (CC50>200 μM), resulting in SI values of>21.9,>14.5 and>121.9, respectively. Compound 4 showed the highest potency with an SI value twice superior to that determined by the standard drug benznidazole (SI>54.6) against the intracellular amastigotes. These data suggest that lignan 4 can be considered a possible scaffold for designing a new drug candidate for Chagas disease.
Collapse
Affiliation(s)
- Marina de M Gonçalves
- Center for Natural and Human Sciences, Federal University of ABC, 09210-580, Santo Andre, SP, Brazil
| | - Carlos H Totini
- Center for Natural and Human Sciences, Federal University of ABC, 09210-580, Santo Andre, SP, Brazil
| | | | | | - Guilherme M Antar
- Department of Agricultural and Biological Sciences, Federal University of Espirito Santo, 29932-540, São Matheus, ES, Brazil
| | | | - João Henrique G Lago
- Center for Natural and Human Sciences, Federal University of ABC, 09210-580, Santo Andre, SP, Brazil
| |
Collapse
|
13
|
Kann S, Concha G, Frickmann H, Hagen RM, Warnke P, Molitor E, Hoerauf A, Backhaus J. Chagas Disease: Comparison of Therapy with Nifurtimox and Benznidazole in Indigenous Communities in Colombia. J Clin Med 2024; 13:2565. [PMID: 38731093 PMCID: PMC11084551 DOI: 10.3390/jcm13092565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Background: For indigenous people in Colombia, high infection rates with Chagas disease (CD) are known. Methods: In 2018 and 2020, nine villages were screened for CD. CD-positive patients could enter a drug observed treatment. While, in 2018, Benznidazole (BNZ) was provided as the first-line drug by the government, nifurtimox (NFX) was administered in 2020. Results: Of 121 individuals treated with BNZ, 79 (65%) suffered from at least one adverse event (AE). Of 115 treated with NFX, at least one AE occurred in 96 (84%) patients. In 69% of BNZ cases, the side effects did not last longer than one day; this applied to 31% of NFX cases. Excluding extreme outlier values, average duration of AEs differed highly significantly: BNZ (M = 0.7, SD = 1.4) and NFX (M = 1.7, SD = 1.5, p < 0.001). Using an intensity scale, AEs were highly significantly more severe for NFX (M = 2.1, SD = 0.58) compared to BZN (M = 1.1, SD = 0.38), p < 0.001. When analyzing the duration in relation to the intensity, the burden of AEs caused by NFX was significantly more pronounced. Dropouts (n = 2) due to AEs were in the NFX-group only. Conclusions: Side effects caused by BNZ were significantly fewer, as well as milder, shorter in duration, and more easily treatable, compared to NFX.
Collapse
Affiliation(s)
- Simone Kann
- Department of Microbiology and Hospital Hygiene, Bundeswehr Central Hospital Koblenz, 56070 Koblenz, Germany;
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany; (E.M.); (A.H.)
| | - Gustavo Concha
- Organization Wiwa Yugumaiun Bunkauanarrua Tayrona (OWYBT), Department Health Advocacy, Valledupar 2000001, Colombia;
| | - Hagen Frickmann
- Department of Microbiology and Hospital Hygiene, Bundeswehr Hospital Hamburg, 20359 Hamburg, Germany;
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, 18057 Rostock, Germany;
| | - Ralf Matthias Hagen
- Department of Microbiology and Hospital Hygiene, Bundeswehr Central Hospital Koblenz, 56070 Koblenz, Germany;
| | - Philipp Warnke
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, 18057 Rostock, Germany;
| | - Ernst Molitor
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany; (E.M.); (A.H.)
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53127 Bonn, Germany; (E.M.); (A.H.)
| | - Joy Backhaus
- Statistical Consulting, 97074 Wuerzburg, Germany;
| |
Collapse
|
14
|
Carey SM, O’Neill DM, Conner GB, Sherman J, Rodriguez A, D’Antonio EL. Discovery of Strong 3-Nitro-2-Phenyl- 2H-Chromene Analogues as Antitrypanosomal Agents and Inhibitors of Trypanosoma cruzi Glucokinase. Int J Mol Sci 2024; 25:4319. [PMID: 38673904 PMCID: PMC11050443 DOI: 10.3390/ijms25084319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Chagas disease is one of the world's neglected tropical diseases, caused by the human pathogenic protozoan parasite Trypanosoma cruzi. There is currently a lack of effective and tolerable clinically available therapeutics to treat this life-threatening illness and the discovery of modern alternative options is an urgent matter. T. cruzi glucokinase (TcGlcK) is a potential drug target because its product, d-glucose-6-phosphate, serves as a key metabolite in the pentose phosphate pathway, glycolysis, and gluconeogenesis. In 2019, we identified a novel cluster of TcGlcK inhibitors that also exhibited anti-T. cruzi efficacy called the 3-nitro-2-phenyl-2H-chromene analogues. This was achieved by performing a target-based high-throughput screening (HTS) campaign of 13,040 compounds. The selection criteria were based on first determining which compounds strongly inhibited TcGlcK in a primary screen, followed by establishing on-target confirmed hits from a confirmatory assay. Compounds that exhibited notable in vitro trypanocidal activity over the T. cruzi infective form (trypomastigotes and intracellular amastigotes) co-cultured in NIH-3T3 mammalian host cells, as well as having revealed low NIH-3T3 cytotoxicity, were further considered. Compounds GLK2-003 and GLK2-004 were determined to inhibit TcGlcK quite well with IC50 values of 6.1 µM and 4.8 µM, respectively. Illuminated by these findings, we herein screened a small compound library consisting of thirteen commercially available 3-nitro-2-phenyl-2H-chromene analogues, two of which were GLK2-003 and GLK2-004 (compounds 1 and 9, respectively). Twelve of these compounds had a one-point change from the chemical structure of GLK2-003. The analogues were run through a similar primary screening and confirmatory assay protocol to our previous HTS campaign. Subsequently, three in vitro biological assays were performed where compounds were screened against (a) T. cruzi (Tulahuen strain) infective form co-cultured within NIH-3T3 cells, (b) T. brucei brucei (427 strain) bloodstream form, and (c) NIH-3T3 host cells alone. We report on the TcGlcK inhibitor constant determinations, mode of enzyme inhibition, in vitro antitrypanosomal IC50 determinations, and an assessment of structure-activity relationships. Our results reveal that the 3-nitro-2-phenyl-2H-chromene scaffold holds promise and can be further optimized for both Chagas disease and human African trypanosomiasis early-stage drug discovery research.
Collapse
Affiliation(s)
- Shane M. Carey
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Destiny M. O’Neill
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Garrett B. Conner
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| | - Julian Sherman
- Department of Microbiology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA (A.R.)
| | - Ana Rodriguez
- Department of Microbiology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA (A.R.)
| | - Edward L. D’Antonio
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA
| |
Collapse
|
15
|
Lapierre TJWJD, Farago DN, de Moura Lodi Cruz MGF, de Melo Resende D, de Oliveira ACR, Dos Santos BRM, de Oliveira Souza F, Michelan-Duarte S, Chelucci RC, Andricopulo AD, Ferreira LLG, Pilau EJ, Murta SMF, de Oliveira Rezende Júnior C. Evaluation and discovery of novel benzothiazole derivatives as promising hits against Leishmania infantum. Chem Biol Drug Des 2024; 103:e14525. [PMID: 38627214 DOI: 10.1111/cbdd.14525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/23/2024] [Accepted: 04/05/2024] [Indexed: 04/19/2024]
Abstract
An early exploration of the benzothiazole class against two kinetoplastid parasites, Leishmania infantum and Trypanosoma cruzi, has been performed after the identification of a benzothiazole derivative as a suitable antileishmanial initial hit. The first series of derivatives focused on the acyl fragment of its class, evaluating diverse linear and cyclic, alkyl and aromatic substituents, and identified two other potent compounds, the phenyl and cyclohexyl derivatives. Subsequently, new compounds were designed to assess the impact of the presence of diverse substituents on the benzothiazole ring or the replacement of the endocyclic sulfur by other heteroatoms. All compounds showed relatively low cytotoxicity, resulting in decent selectivity indexes for the most active compounds. Ultimately, the in vitro ADME properties of these compounds were assessed, revealing a satisfying water solubility, gastrointestinal permeability, despite their low metabolic stability and high lipophilicity. Consequently, compounds 5 and 6 were identified as promising hits for further hit-to-lead exploration within this benzothiazole class against L. infantum, thus providing promising starting points for the development of antileishmanial candidates.
Collapse
Affiliation(s)
| | - Danilo Nascimento Farago
- Laboratório de Síntese de Candidatos a Fármacos, Instituto de Química, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | | | - Daniela de Melo Resende
- Grupo de Genômica Funcional de Parasitos, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ Minas), Belo Horizonte, Minas Gerais, Brazil
| | - Adriane Cristina Rosa de Oliveira
- Laboratório de Síntese de Candidatos a Fármacos, Instituto de Química, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | - Brenda Rosa Macedo Dos Santos
- Laboratório de Síntese de Candidatos a Fármacos, Instituto de Química, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | - Felipe de Oliveira Souza
- Laboratório de Biomoléculas e Espectrometria de Massas (LaBioMass), Universidade Estadual de Maringá (UEM), Maringá, Paraná, Brazil
| | - Simone Michelan-Duarte
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos, São Paulo, Brazil
| | - Rafael C Chelucci
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos, São Paulo, Brazil
| | - Adriano D Andricopulo
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos, São Paulo, Brazil
| | - Leonardo L G Ferreira
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos, São Paulo, Brazil
| | - Eduardo Jorge Pilau
- Laboratório de Biomoléculas e Espectrometria de Massas (LaBioMass), Universidade Estadual de Maringá (UEM), Maringá, Paraná, Brazil
| | - Silvane Maria Fonseca Murta
- Grupo de Genômica Funcional de Parasitos, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ Minas), Belo Horizonte, Minas Gerais, Brazil
| | - Celso de Oliveira Rezende Júnior
- Laboratório de Síntese de Candidatos a Fármacos, Instituto de Química, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| |
Collapse
|
16
|
Selener MG, Borgo J, Sarratea MB, Delfino MA, Laurella LC, Cerny N, Gomez J, Coll M, Malchiodi EL, Bivona AE, Barrera P, Redko FC, Catalán CAN, Alberti AS, Sülsen VP. Trypanocidal and Anti-Inflammatory Effects of Three ent-Kaurane Diterpenoids from Gymnocoronis spilanthoides var. subcordata (Asteraceae). Pharmaceutics 2024; 16:415. [PMID: 38543309 PMCID: PMC10975076 DOI: 10.3390/pharmaceutics16030415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 01/03/2025] Open
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, affects 6-7 million people worldwide. The dichloromethane extract obtained from the aerial parts of Gymnocoronis spilanthoides var subcordata showed trypanocidal activity in vitro. The fractionation of the dewaxed organic extract via column chromatography led to the isolation of three diterpenoids: ent-9α,11α-dihydroxy-15-oxo-kaur-16-en-19-oic acid or adenostemmoic acid B, (16R)-ent-11α-hydroxy-15-oxokauran-19-oic acid and ent-11α-hydroxy-15-oxo-kaur-16-en-19-oic acid. These compounds showed IC50 values of 10.6, 15.9 and 4.8 µM against T. cruzi epimastigotes, respectively. When tested against amastigotes, the diterpenoids afforded IC50 values of 6.1, 19.5 and 60.6 µM, respectively. The cytotoxicity of the compounds was tested on mammalian cells using an MTT assay, resulting in CC50s of 321.8, 23.3 and 14.8 µM, respectively. The effect of adenostemmoic acid B on T. cruzi was examined at the ultrastructural level using transmission microscopy. Treatment with 20 μM for 48 h stimulated the formation of abnormal cytosolic membranous structures in the parasite. This compound also showed an anti-inflammatory effect in murine macrophages stimulated with LPS and other TLR agonists. Treatment of macrophages with adenostemmoic acid B was able to reduce TNF secretion and nitric oxide production, while increasing IL-10 production. The combination of adenostemmoic acid B with benznidazole resulted in greater inhibition of NF-kB and a decrease in nitrite concentration. The administration of adenostemmoic acid B to mice infected with trypomastigotes of T. cruzi at the dose of 1 mg/kg/day for five days produced a significant decrease in parasitemia levels and weight loss. Treatment with the association with benznidazole increased the survival time of the animals. In view of these results, adenostemmoic acid B could be considered a promising candidate for further studies in the search for new treatments for Chagas disease.
Collapse
Affiliation(s)
- Mariana G. Selener
- Cátedra de Farmacognosia, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina; (M.G.S.); (J.B.); (L.C.L.); (F.C.R.)
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), CONICET-Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina
| | - Jimena Borgo
- Cátedra de Farmacognosia, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina; (M.G.S.); (J.B.); (L.C.L.); (F.C.R.)
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), CONICET-Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina
| | - Maria Belen Sarratea
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina; (M.B.S.); (E.L.M.); (A.E.B.)
| | - Maria Alicia Delfino
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina;
| | - Laura C. Laurella
- Cátedra de Farmacognosia, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina; (M.G.S.); (J.B.); (L.C.L.); (F.C.R.)
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), CONICET-Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina
| | - Natacha Cerny
- Departamento de Microbiología, Parasitología e Inmunología-IMPAM (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires C1121ABG, Argentina;
| | - Jessica Gomez
- Facultad de Ciencias Médicas, Instituto de Histología y Embriología “Dr. Mario H. Burgos” (IHEM), Universidad Nacional de Cuyo-CONICET, CC 56, Mendoza 5500, Argentina; (J.G.); (M.C.); (P.B.)
| | - Mauro Coll
- Facultad de Ciencias Médicas, Instituto de Histología y Embriología “Dr. Mario H. Burgos” (IHEM), Universidad Nacional de Cuyo-CONICET, CC 56, Mendoza 5500, Argentina; (J.G.); (M.C.); (P.B.)
| | - Emilio L. Malchiodi
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina; (M.B.S.); (E.L.M.); (A.E.B.)
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina;
| | - Augusto E. Bivona
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina; (M.B.S.); (E.L.M.); (A.E.B.)
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina;
| | - Patricia Barrera
- Facultad de Ciencias Médicas, Instituto de Histología y Embriología “Dr. Mario H. Burgos” (IHEM), Universidad Nacional de Cuyo-CONICET, CC 56, Mendoza 5500, Argentina; (J.G.); (M.C.); (P.B.)
| | - Flavia C. Redko
- Cátedra de Farmacognosia, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina; (M.G.S.); (J.B.); (L.C.L.); (F.C.R.)
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), CONICET-Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina
| | - César A. N. Catalán
- Instituto de Química Orgánica, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Ayacucho 471, San Miguel de Tucumán T4000INI, Argentina;
| | - Andrés Sánchez Alberti
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-Universidad de Buenos Aires, Junín 956, Piso 4, Buenos Aires C1113AAD, Argentina; (M.B.S.); (E.L.M.); (A.E.B.)
- Departamento de Microbiología, Parasitología e Inmunología-IMPAM (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, Buenos Aires C1121ABG, Argentina;
| | - Valeria P. Sülsen
- Cátedra de Farmacognosia, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina; (M.G.S.); (J.B.); (L.C.L.); (F.C.R.)
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), CONICET-Universidad de Buenos Aires, Junín 956, Piso 2, Buenos Aires C1113AAD, Argentina
| |
Collapse
|
17
|
Carey SM, Kearns SP, Millington ME, Buechner GS, Alvarez BE, Daneshian L, Abiskaroon B, Chruszcz M, D'Antonio EL. At the outer part of the active site in Trypanosoma cruzi glucokinase: The role of phenylalanine 337. Biochimie 2024; 218:8-19. [PMID: 37741546 DOI: 10.1016/j.biochi.2023.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/10/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023]
Abstract
The hole mutagenesis approach was used to interrogate the importance of F337 in Trypanosoma cruzi glucokinase (TcGlcK) in order to understand the complete set of binding interactions that are made by d-glucosamine analogue inhibitors containing aromatic tail groups that can extend to the outer part of the active site. An interesting inhibitor of this analogue class includes 2-N-carboxybenzyl-2-deoxy-d-glucosamine (CBZ-GlcN), which exhibits strong TcGlcK binding with a Ki of 710 nM. The residue F337 is found at the outer part of the active site that stems from the second protein subunit of the homodimeric assembly. In this study, F337 was changed to leucine and alanine so as to diminish phenylalanine's side chain size and attenuate intermolecular interactions in this region of the binding cavity. Results from enzyme - inhibitor assays revealed that the phenyl group of F337 made dominant hydrophobic interactions with the phenyl group of CBZ-GlcN as opposed to π - π stacking interactions. Moreover, enzymatic activity assays and X-ray crystallographic experiments indicated that each of these site-directed mutants primarily retained their activity and had high structural similarity of their protein fold. A computed structure model of T. cruzi hexokinase (TcHxK), which was produced by the artificial intelligence system AlphaFold, was compared to an X-ray crystal structure of TcGlcK. Our structural analysis revealed that TcHxK lacked an F337 counterpart residue and probably exists in the monomeric form. We proposed that the d-glucosamine analogue inhibitors that are structurally similar to CBZ-GlcN may not bind as strongly in TcHxK as they do in TcGlcK because of absent van der Waals contact from residue side chains.
Collapse
Affiliation(s)
- Shane M Carey
- Department of Natural Sciences, University of South Carolina Beaufort, Bluffton, SC 29909, USA
| | - Sean P Kearns
- Department of Natural Sciences, University of South Carolina Beaufort, Bluffton, SC 29909, USA
| | - Matthew E Millington
- Department of Natural Sciences, University of South Carolina Beaufort, Bluffton, SC 29909, USA
| | - Gregory S Buechner
- Department of Natural Sciences, University of South Carolina Beaufort, Bluffton, SC 29909, USA
| | - Beda E Alvarez
- Department of Natural Sciences, University of South Carolina Beaufort, Bluffton, SC 29909, USA
| | - Leily Daneshian
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Brendan Abiskaroon
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Maksymilian Chruszcz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Edward L D'Antonio
- Department of Natural Sciences, University of South Carolina Beaufort, Bluffton, SC 29909, USA.
| |
Collapse
|
18
|
Dube NP, Thatyana M, Mokgalaka-Fleischmann NS, Mansour AM, Tembu VJ, Manicum ALE. Review on the Applications of Selected Metal-Based Complexes on Infectious Diseases. Molecules 2024; 29:406. [PMID: 38257319 PMCID: PMC10819944 DOI: 10.3390/molecules29020406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Fatalities caused by infectious diseases (i.e., diseases caused by parasite, bacteria, and viruses) have become reinstated as a major public health threat globally. Factors such as antimicrobial resistance and viral complications are the key contributors to the death numbers. As a result, new compounds with structural diversity classes are critical for controlling the virulence of pathogens that are multi-drug resistant. Derivatization of bio-active organic molecules with organometallic synthons is a promising strategy for modifying the inherent and enhanced properties of biomolecules. Due to their redox chemistry, bioactivity, and structural diversity, organometallic moieties make excellent candidates for lead structures in drug development. Furthermore, organometallic compounds open an array of potential in therapy that existing organic molecules lack, i.e., their ability to fulfill drug availability and resolve the frequent succumbing of organic molecules to drug resistance. Additionally, metal complexes have the potential towards metal-specific modes of action, preventing bacteria from developing resistance mechanisms. This review's main contribution is to provide a thorough account of the biological efficacy (in vitro and in vitro) of metal-based complexes against infectious diseases. This resource can also be utilized in conjunction with corresponding journals on metal-based complexes investigated against infectious diseases.
Collapse
Affiliation(s)
- Nondumiso P. Dube
- Department of Chemistry, Tshwane University of Technology, 175 Nelson Mandela Drive, Private Bag X680, Pretoria 0001, South Africa; (N.P.D.); (M.T.); (N.S.M.-F.); (V.J.T.)
| | - Maxwell Thatyana
- Department of Chemistry, Tshwane University of Technology, 175 Nelson Mandela Drive, Private Bag X680, Pretoria 0001, South Africa; (N.P.D.); (M.T.); (N.S.M.-F.); (V.J.T.)
| | - Ntebogeng S. Mokgalaka-Fleischmann
- Department of Chemistry, Tshwane University of Technology, 175 Nelson Mandela Drive, Private Bag X680, Pretoria 0001, South Africa; (N.P.D.); (M.T.); (N.S.M.-F.); (V.J.T.)
| | - Ahmed M. Mansour
- Department of Chemistry, United Arab Emirates University, Al-Ain 15551, United Arab Emirates;
| | - Vuyelwa J. Tembu
- Department of Chemistry, Tshwane University of Technology, 175 Nelson Mandela Drive, Private Bag X680, Pretoria 0001, South Africa; (N.P.D.); (M.T.); (N.S.M.-F.); (V.J.T.)
| | - Amanda-Lee E. Manicum
- Department of Chemistry, Tshwane University of Technology, 175 Nelson Mandela Drive, Private Bag X680, Pretoria 0001, South Africa; (N.P.D.); (M.T.); (N.S.M.-F.); (V.J.T.)
| |
Collapse
|
19
|
González S, Wall RJ, Thomas J, Braillard S, Brunori G, Díaz IC, Cantizani J, Carvalho S, Castañeda Casado P, Chatelain E, Cotillo I, Fiandor JM, Francisco AF, Grimsditch D, Keenan M, Kelly JM, Kessler A, Luise C, Lyon JJ, MacLean L, Marco M, Martin JJ, Martinez MS, Paterson C, Read KD, Santos-Villarejo A, Zuccotto F, Wyllie S, Miles TJ, De Rycker M. Short-course combination treatment for experimental chronic Chagas disease. Sci Transl Med 2023; 15:eadg8105. [PMID: 38091410 PMCID: PMC7615676 DOI: 10.1126/scitranslmed.adg8105] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023]
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, affects millions of people in the Americas and across the world, leading to considerable morbidity and mortality. Current treatment options, benznidazole (BNZ) and nifurtimox, offer limited efficacy and often lead to adverse side effects because of long treatment durations. Better treatment options are therefore urgently required. Here, we describe a pyrrolopyrimidine series, identified through phenotypic screening, that offers an opportunity to improve on current treatments. In vitro cell-based washout assays demonstrate that compounds in the series are incapable of killing all parasites; however, combining these pyrrolopyrimidines with a subefficacious dose of BNZ can clear all parasites in vitro after 5 days. These findings were replicated in a clinically predictive in vivo model of chronic Chagas disease, where 5 days of treatment with the combination was sufficient to prevent parasite relapse. Comprehensive mechanism of action studies, supported by ligand-structure modeling, show that compounds from this pyrrolopyrimidine series inhibit the Qi active site of T. cruzi cytochrome b, part of the cytochrome bc1 complex of the electron transport chain. Knowledge of the molecular target enabled a cascade of assays to be assembled to evaluate selectivity over the human cytochrome b homolog. As a result, a highly selective and efficacious lead compound was identified. The combination of our lead compound with BNZ rapidly clears T. cruzi parasites, both in vitro and in vivo, and shows great potential to overcome key issues associated with currently available treatments.
Collapse
Affiliation(s)
- Silvia González
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Richard J. Wall
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - John Thomas
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | | | | | - Juan Cantizani
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Sandra Carvalho
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | | | - Ignacio Cotillo
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Jose M. Fiandor
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | | | | | | | - John M. Kelly
- London School for Hygiene and Tropical Medicine, London, UK
| | - Albane Kessler
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Chiara Luise
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | - Lorna MacLean
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Maria Marco
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - J. Julio Martin
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | | | - Christy Paterson
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Kevin D. Read
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | - Fabio Zuccotto
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Susan Wyllie
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Tim J. Miles
- Global Health Medicines R&D, GSK, Tres Cantos, Madrid, Spain
| | - Manu De Rycker
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| |
Collapse
|
20
|
Bosch-Navarrete C, Pérez-Moreno G, Annang F, Diaz-Gonzalez R, García-Hernández R, Rocha H, Gamarro F, Cordón-Obras C, Navarro M, Rodriguez A, Genilloud O, Reyes F, Vicente F, Ruiz-Pérez LM, González-Pacanowska D. Strasseriolides display in vitro and in vivo activity against trypanosomal parasites and cause morphological and size defects in Trypanosoma cruzi. PLoS Negl Trop Dis 2023; 17:e0011592. [PMID: 37713416 PMCID: PMC10529594 DOI: 10.1371/journal.pntd.0011592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 09/27/2023] [Accepted: 08/14/2023] [Indexed: 09/17/2023] Open
Abstract
Neglected diseases caused by kinetoplastid parasites are a health burden in tropical and subtropical countries. The need to create safe and effective medicines to improve treatment remains a priority. Microbial natural products are a source of chemical diversity that provides a valuable approach for identifying new drug candidates. We recently reported the discovery and bioassay-guided isolation of a novel family of macrolides with antiplasmodial activity. The novel family of four potent antimalarial macrolides, strasseriolides A-D, was isolated from cultures of Strasseria geniculata CF-247251, a fungal strain obtained from plant tissues. In the present study, we analyze these strasseriolides for activity against kinetoplastid protozoan parasites, namely, Trypanosoma brucei brucei, Leishmania donovani and Trypanosoma cruzi. Compounds exhibited mostly low activities against T. b. brucei, yet notable growth inhibition and selectivity were observed for strasseriolides C and D in the clinically relevant intracellular T. cruzi and L. donovani amastigotes with EC50 values in the low micromolar range. Compound C is fast-acting and active against both intracellular and trypomastigote forms of T. cruzi. While cell cycle defects were not identified, prominent morphological changes were visualized by differential interference contrast microscopy and smaller and rounded parasites were visualized upon exposure to strasseriolide C. Moreover, compound C lowers parasitaemia in vivo in acute models of infection of Chagas disease. Hence, strasseriolide C is a novel natural product active against different forms of T. cruzi in vitro and in vivo. The study provides an avenue for blocking infection of new cells, a strategy that could additionally contribute to avoid treatment failure.
Collapse
Affiliation(s)
- Cristina Bosch-Navarrete
- Instituto de Parasitología y Biomedicina "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Guiomar Pérez-Moreno
- Instituto de Parasitología y Biomedicina "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Frederick Annang
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Rosario Diaz-Gonzalez
- Instituto de Parasitología y Biomedicina "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Raquel García-Hernández
- Instituto de Parasitología y Biomedicina "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Hedy Rocha
- Department of Microbiology, Core Anti-infectives, New York University School of Medicine, New York, New York, United States of America
| | - Francisco Gamarro
- Instituto de Parasitología y Biomedicina "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Carlos Cordón-Obras
- Instituto de Parasitología y Biomedicina "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Miguel Navarro
- Instituto de Parasitología y Biomedicina "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Ana Rodriguez
- Department of Microbiology, Core Anti-infectives, New York University School of Medicine, New York, New York, United States of America
| | - Olga Genilloud
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Fernando Reyes
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Francisca Vicente
- Fundación MEDINA, Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Luis M. Ruiz-Pérez
- Instituto de Parasitología y Biomedicina "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Parque Tecnológico Ciencias de la Salud, Granada, Spain
| | - Dolores González-Pacanowska
- Instituto de Parasitología y Biomedicina "López-Neyra", Consejo Superior de Investigaciones Científicas (CSIC), Parque Tecnológico Ciencias de la Salud, Granada, Spain
| |
Collapse
|
21
|
Thomas M, McGonagle K, Rowland P, Robinson DA, Dodd PG, Camino-Díaz I, Campbell L, Cantizani J, Castañeda P, Conn D, Craggs PD, Edwards D, Ferguson L, Fosberry A, Frame L, Goswami P, Hu X, Korczynska J, MacLean L, Martin J, Mutter N, Osuna-Cabello M, Paterson C, Peña I, Pinto EG, Pont C, Riley J, Shishikura Y, Simeons FRC, Stojanovski L, Thomas J, Wrobel K, Young RJ, Zmuda F, Zuccotto F, Read KD, Gilbert IH, Marco M, Miles TJ, Manzano P, De Rycker M. Structure-Guided Design and Synthesis of a Pyridazinone Series of Trypanosoma cruzi Proteasome Inhibitors. J Med Chem 2023; 66:10413-10431. [PMID: 37506194 PMCID: PMC10424187 DOI: 10.1021/acs.jmedchem.3c00582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Indexed: 07/30/2023]
Abstract
There is an urgent need for new treatments for Chagas disease, a parasitic infection which mostly impacts South and Central America. We previously reported on the discovery of GSK3494245/DDD01305143, a preclinical candidate for visceral leishmaniasis which acted through inhibition of the Leishmania proteasome. A related analogue, active against Trypanosoma cruzi, showed suboptimal efficacy in an animal model of Chagas disease, so alternative proteasome inhibitors were investigated. Screening a library of phenotypically active analogues against the T. cruzi proteasome identified an active, selective pyridazinone, the development of which is described herein. We obtained a cryo-EM co-structure of proteasome and a key inhibitor and used this to drive optimization of the compounds. Alongside this, optimization of the absorption, distribution, metabolism, and excretion (ADME) properties afforded a suitable compound for mouse efficacy studies. The outcome of these studies is discussed, alongside future plans to further understand the series and its potential to deliver a new treatment for Chagas disease.
Collapse
Affiliation(s)
- Michael
G. Thomas
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Kate McGonagle
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Paul Rowland
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - David A. Robinson
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Peter G. Dodd
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Isabel Camino-Díaz
- GlaxoSmithKline,
Discovery DMPK, IVIVT, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Lorna Campbell
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Juan Cantizani
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Pablo Castañeda
- GlaxoSmithKline,
Discovery DMPK, IVIVT, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Daniel Conn
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Peter D. Craggs
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Darren Edwards
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Liam Ferguson
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Andrew Fosberry
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Laura Frame
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Panchali Goswami
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Xiao Hu
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Justyna Korczynska
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Lorna MacLean
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Julio Martin
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Nicole Mutter
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Maria Osuna-Cabello
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Christy Paterson
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Imanol Peña
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Erika G. Pinto
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Caterina Pont
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Jennifer Riley
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Yoko Shishikura
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Frederick R. C. Simeons
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Laste Stojanovski
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - John Thomas
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Karolina Wrobel
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | | | - Filip Zmuda
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Fabio Zuccotto
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Kevin D. Read
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Ian H. Gilbert
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Maria Marco
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Timothy J. Miles
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Pilar Manzano
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Manu De Rycker
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| |
Collapse
|
22
|
Vieira da Silva Torchelsen FK, Fernandes Pedrosa TC, Rodrigues MP, de Aguiar AR, de Oliveira FM, Amarante GW, Sales-Junior PA, Branquinho RT, Gomes da Silva SP, Talvani A, Fonseca Murta SM, Martins FT, Braun RL, Teixeira RR, Furtado Mosqueira VC, Lana MD. Novel diamides inspired by protein kinase inhibitors as anti- Trypanosoma cruzi agents: in vitro and in vivo evaluations. Future Med Chem 2023; 15:1469-1489. [PMID: 37650735 DOI: 10.4155/fmc-2023-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
Background: Chagas disease is a life-threatening illness caused by Trypanosoma cruzi. The involvement of serine-/arginine-rich protein kinase in the T. cruzi life cycle is significant. Aims: To synthesize, characterize and evaluate the trypanocidal activity of diamides inspired by kinase inhibitor, SRPIN340. Material & Methods: Synthesis using a three-step process and characterization by infrared, nuclear magnetic resonance and high-resolution mass spectrometry were conducted. The selectivity index was obtained by the ratio of CC50/IC50 in two in vitro models. The most active compound, 3j, was evaluated using in vitro cytokine assays and assessing in vivo trypanocidal activity. Results: 3j activity in the macrophage J774 lineage showed an anti-inflammatory profile, and mice showed significantly reduced parasitemia and morbidity at low compound dosages. Conclusion: Novel diamide is active against T. cruzi in vitro and in vivo.
Collapse
Affiliation(s)
| | - Tamiles Caroline Fernandes Pedrosa
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | | | - Alex Ramos de Aguiar
- Departamento de Química, Universidade Federal de Viçosa, Viçosa, Minas Gerais, 30130-171, Brazil
| | | | - Giovanni Wilson Amarante
- Departamento de Química, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, 36036-900, Brazil
| | | | - Renata Tupinambá Branquinho
- Programa de Pós-Graduação em Ciências Farmacêuticas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Sirlaine Pio Gomes da Silva
- Programa de pós-graduação em Saúde e Nutrição, Escola de Nutrição, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - André Talvani
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
- Programa de pós-graduação em Saúde e Nutrição, Escola de Nutrição, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | | | - Felipe Terra Martins
- Departamento de Química, Universidade Federal de Goiás, Goiânia, Goiás, 74001-970, Brazil
| | - Rodrigo Ligabue Braun
- Departamento de Ciências Farmacêuticas, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, 90050-170, Brazil
| | - Róbson Ricardo Teixeira
- Departamento de Química, Universidade Federal de Viçosa, Viçosa, Minas Gerais, 30130-171, Brazil
| | - Vanessa Carla Furtado Mosqueira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Marta de Lana
- Programa de Pós-Graduação em Ciências Farmacêuticas, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| |
Collapse
|
23
|
Nunes EAC, da Silva MC, Cardoso MH, Preza SLE, de Oliveira LS, Frihling BEF, Charneau SO, Grellier P, Franco OL, Migliolo L. Anti-Protozoan Activities of Polar Fish-Derived Polyalanine Synthetic Peptides. Mar Drugs 2023; 21:434. [PMID: 37623715 PMCID: PMC10456387 DOI: 10.3390/md21080434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023] Open
Abstract
Chagas disease, sleeping sickness and malaria are infectious diseases caused by protozoan parasites that kill millions of people worldwide. Here, we performed in vitro assays of Pa-MAP, Pa-MAP1.9, and Pa-MAP2 synthetic polyalanine peptides derived from the polar fish Pleuronectes americanus toward Trypanosoma cruzi, T. brucei gambiense and Plasmodium falciparum activities. We demonstrated that the peptides Pa-MAP1.9 and Pa-MAP2 were effective to inhibit T. brucei growth. In addition, structural analyses using molecular dynamics (MD) studies showed that Pa-MAP2 penetrates deeper into the membrane and interacts more with phospholipids than Pa-MAP1.9, corroborating the previous in vitro results showing that Pa-MAP1.9 acts within the cell, while Pa-MAP2 acts via membrane lysis. In conclusion, polyalanine Pa-MAP1.9 and Pa-MAP2 presented activity against bloodstream forms of T. b. gambiense, thus encouraging further studies on the application of these peptides as a treatment for sleeping sickness.
Collapse
Affiliation(s)
| | - Maria Cláudia da Silva
- S-Inova Biotech, Graduate Program in Biotechnology, Dom Bosco Catholic University, Campo Grande 79117-900, Brazil
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Sao Paulo 14040-900, Brazil
- Laboratory of Research in Virology and Immunology, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio 57020-600, Brazil
| | - Marlon Henrique Cardoso
- S-Inova Biotech, Graduate Program in Biotechnology, Dom Bosco Catholic University, Campo Grande 79117-900, Brazil
- Center for Proteomics and Biochemical Analysis, Catholic University of Brasília, Brasilia 71966-700, Brazil
| | | | - Lucas Silva de Oliveira
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasilia 73345-010, Brazil
- UMR 7245 Molécules de Communication et Adaptation des Micro-Organismes, Muséum National d'Histoire Naturelle, CNRS, 75005 Paris, France
- Graduate Program in Molecular Pathology, University of Brasilia, Brasilia 73345-010, Brazil
| | | | - Sébastien Olivier Charneau
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Brasilia 73345-010, Brazil
- Graduate Program in Molecular Pathology, University of Brasilia, Brasilia 73345-010, Brazil
| | - Philippe Grellier
- UMR 7245 Molécules de Communication et Adaptation des Micro-Organismes, Muséum National d'Histoire Naturelle, CNRS, 75005 Paris, France
| | - Octávio Luiz Franco
- S-Inova Biotech, Graduate Program in Biotechnology, Dom Bosco Catholic University, Campo Grande 79117-900, Brazil
- Center for Proteomics and Biochemical Analysis, Catholic University of Brasília, Brasilia 71966-700, Brazil
| | - Ludovico Migliolo
- Postgraduate Program in Biochemistry, Federal University of Rio Grande do Norte, Natal 59078-900, Brazil
- S-Inova Biotech, Graduate Program in Biotechnology, Dom Bosco Catholic University, Campo Grande 79117-900, Brazil
- Postgraduate Program in Cellular and Molecular Biology, Federal University of Paraíba, João Pessoa 58051-900, Brazil
| |
Collapse
|
24
|
Laureano de Souza M, Lapierre TJWJD, Vitor de Lima Marques G, Ferraz WR, Penteado AB, Henrique Goulart Trossini G, Murta SMF, de Oliveira RB, de Oliveira Rezende C, Ferreira RS. Molecular targets for Chagas disease: validation, challenges and lead compounds for widely exploited targets. Expert Opin Ther Targets 2023; 27:911-925. [PMID: 37772733 DOI: 10.1080/14728222.2023.2264512] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/24/2023] [Indexed: 09/30/2023]
Abstract
INTRODUCTION Chagas disease (CD) imposes social and economic burdens, yet the available treatments have limited efficacy in the disease's chronic phase and cause serious adverse effects. To address this challenge, target-based approaches are a possible strategy to develop new, safe, and active treatments for both phases of the disease. AREAS COVERED This review delves into target-based approaches applied to CD drug discovery, emphasizing the studies from the last five years. We highlight the proteins cruzain (CZ), trypanothione reductase (TR), sterol 14 α-demethylase (CPY51), iron superoxide dismutase (Fe-SOD), proteasome, cytochrome b (Cytb), and cleavage and polyadenylation specificity factor 3 (CPSF3), chosen based on their biological and chemical validation as drug targets. For each, we discuss its biological relevance and validation as a target, currently related challenges, and the status of the most promising inhibitors. EXPERT OPINION Target-based approaches toward developing potential CD therapeutics have yielded promising leads in recent years. We expect a significant advance in this field in the next decade, fueled by the new options for Trypanosoma cruzi genetic manipulation that arose in the past decade, combined with recent advances in computational chemistry and chemical biology.
Collapse
Affiliation(s)
- Mariana Laureano de Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Gabriel Vitor de Lima Marques
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Witor Ribeiro Ferraz
- Departamento de Farmacia, Faculdade de Ciencias Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - André Berndt Penteado
- Departamento de Farmacia, Faculdade de Ciencias Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Renata Barbosa de Oliveira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Rafaela Salgado Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
25
|
Gabaldón-Figueira JC, Martinez-Peinado N, Escabia E, Ros-Lucas A, Chatelain E, Scandale I, Gascon J, Pinazo MJ, Alonso-Padilla J. State-of-the-Art in the Drug Discovery Pathway for Chagas Disease: A Framework for Drug Development and Target Validation. Res Rep Trop Med 2023; 14:1-19. [PMID: 37337597 PMCID: PMC10277022 DOI: 10.2147/rrtm.s415273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/03/2023] [Indexed: 06/21/2023] Open
Abstract
Chagas disease is the most important protozoan infection in the Americas, and constitutes a significant public health concern throughout the world. Development of new medications against its etiologic agent, Trypanosoma cruzi, has been traditionally slow and difficult, lagging in comparison with diseases caused by other kinetoplastid parasites. Among the factors that explain this are the incompletely understood mechanisms of pathogenesis of T. cruzi infection and its complex set of interactions with the host in the chronic stage of the disease. These demand the performance of a variety of in vitro and in vivo assays as part of any drug development effort. In this review, we discuss recent breakthroughs in the understanding of the parasite's life cycle and their implications in the search for new chemotherapeutics. For this, we present a framework to guide drug discovery efforts against Chagas disease, considering state-of-the-art preclinical models and recently developed tools for the identification and validation of molecular targets.
Collapse
Affiliation(s)
| | - Nieves Martinez-Peinado
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
| | - Elisa Escabia
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
| | - Albert Ros-Lucas
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - Eric Chatelain
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Ivan Scandale
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Joaquim Gascon
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - María-Jesús Pinazo
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| |
Collapse
|
26
|
Singh VK, Kumar A. Secondary metabolites from endophytic fungi: Production, methods of analysis, and diverse pharmaceutical potential. Symbiosis 2023; 90:1-15. [PMID: 37360552 PMCID: PMC10249938 DOI: 10.1007/s13199-023-00925-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023]
Abstract
The synthesis of secondary metabolites is a constantly functioning metabolic pathway in all living systems. Secondary metabolites can be broken down into numerous classes, including alkaloids, coumarins, flavonoids, lignans, saponins, terpenes, quinones, xanthones, and others. However, animals lack the routes of synthesis of these compounds, while plants, fungi, and bacteria all synthesize them. The primary function of bioactive metabolites (BM) synthesized from endophytic fungi (EF) is to make the host plants resistant to pathogens. EF is a group of fungal communities that colonize host tissues' intracellular or intercellular spaces. EF serves as a storehouse of the above-mentioned bioactive metabolites, providing beneficial effects to their hosts. BM of EF could be promising candidates for anti-cancer, anti-malarial, anti-tuberculosis, antiviral, anti-inflammatory, etc. because EF is regarded as an unexploited and untapped source of novel BM for effective drug candidates. Due to the emergence of drug resistance, there is an urgent need to search for new bioactive compounds that combat resistance. This article summarizes the production of BM from EF, high throughput methods for analysis, and their pharmaceutical application. The emphasis is on the diversity of metabolic products from EF, yield, method of purification/characterization, and various functions/activities of EF. Discussed information led to the development of new drugs and food additives that were more effective in the treatment of disease. This review shed light on the pharmacological potential of the fungal bioactive metabolites and emphasizes to exploit them in the future for therapeutic purposes.
Collapse
Affiliation(s)
- Vivek Kumar Singh
- Department of Biotechnology, National Institute of Technology, Raipur (CG), Raipur, 492010 Chhattisgarh India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur (CG), Raipur, 492010 Chhattisgarh India
| |
Collapse
|
27
|
Silva-Oliveira R, Sangenito LS, Reddy A, Velasco-Torrijos T, Santos ALS, Branquinha MH. In Vitro Effects of Aminopyridyl Ligands Complexed to Copper(II) on the Physiology and Interaction Process of Trypanosoma cruzi. Trop Med Infect Dis 2023; 8:tropicalmed8050288. [PMID: 37235336 DOI: 10.3390/tropicalmed8050288] [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: 03/24/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
Chagas disease is derived from the infection by the protozoan Trypanosoma cruzi. In many countries, benznidazole is the only drug approved for clinical use despite several side effects and the emergence of resistant parasite strains. In this context, our group has previously pointed out that two novel aminopyridine derivatives complexed with Cu2+, namely, cis-aquadichloro(N-[4-(hydroxyphenyl)methyl]-2-pyridinemethamino)copper (3a) and its glycosylated ligand cis-dichloro (N-{[4-(2,3,4,6-tetra-O-acetyl-β-D-glucopyranosyloxy)pheny]lmethyl}-2-pyridinemethamino)copper (3b), are effective against T. cruzi trypomastigote forms. With this result in mind, the present work aimed to investigate the effects of both compounds on trypomastigotes physiology and on the interaction process with host cells. Apart from loss of plasma membrane integrity, an increased generation of reactive oxygen species (ROS) and decreased mitochondrial metabolism were observed. Pretreatment of trypomastigotes with these metallodrugs inhibited the association index with LLC-MK2 cells in a typical dose-dependent manner. Both compounds showed low toxicity on mammalian cells (CC50 > 100 µM), and the IC50 values calculated for intracellular amastigotes were determined as 14.4 µM for 3a and 27.1 µM for 3b. This set of results demonstrates the potential of these aminopyridines complexed with Cu2+ as promising candidates for further antitrypanosomal drug development.
Collapse
Affiliation(s)
- Rafaela Silva-Oliveira
- Laboratório de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Leandro S Sangenito
- Laboratório de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro, Nilópolis 26530-060, Brazil
| | - Andrew Reddy
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt 5010, New Zealand
| | - Trinidad Velasco-Torrijos
- Department of Chemistry, Maynooth University, W23VP22 Maynooth, Co. Kildare, Ireland
- The Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23VP22 Maynooth, Co. Kildare, Ireland
| | - André L S Santos
- Laboratório de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Programa de Pós-Graduação em Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| | - Marta H Branquinha
- Laboratório de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
28
|
Pagotti MC, Dias HJ, Candido ACBB, Oliveira TAS, Borges A, Oliveira ND, Lopes CD, Orenha RP, Parreira RLT, Crotti AEM, Magalhães LG. Exploring Synthetic Dihydrobenzofuran and Benzofuran Neolignans as Antiprotozoal Agents against Trypanosoma cruzi. Pharmaceutics 2023; 15:pharmaceutics15030754. [PMID: 36986617 PMCID: PMC10052957 DOI: 10.3390/pharmaceutics15030754] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 03/30/2023] Open
Abstract
Chagas disease is a neglected tropical disease that affects more than 8 million people. Although there are therapies against this disease, the search for new drugs is important because the current treatments show limited effectiveness and high toxicity. In this work, eighteen dihydrobenzofuran-type neolignans (DBNs) and two benzofuran-type neolignans (BNs) were synthesized and evaluated against amastigote forms of two Trypanosoma cruzi strains. The in vitro cytotoxicity and hemolytic activity of the most active compounds were also evaluated and their relationships with T. cruzi tubulin DBNs were investigated by an in silico approach. Four DBNs demonstrated activity against the T. cruzi Tulahuen lac-Z strain (IC50 from 7.96 to 21.12 µM), and DBN 1 exhibited the highest activity against the amastigote forms of the T. cruzi Y strain (IC50 3.26 μM). Compounds 1-4 showed CC50 values higher than antitrypanosomal activities, except for DBN 3. All DBNs with antitrypanosomal activity demonstrated CH50 higher than 100 µM. The in silico results indicated that DBNs 1, 2, and 4 are capable of destabilizing the dynamics of the tubulin-microtubule from the vinca site. These compounds displayed promising in vitro activity against T. cruzi, especially compound 1, and can be considered molecular prototypes for the development of new antiparasitic drugs.
Collapse
Affiliation(s)
- Mariana C Pagotti
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
| | - Herbert J Dias
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
- Goiano Federal Institute of Education, Science, and Technology, Campus Urutaí, Urutaí 75790-000, GO, Brazil
| | - Ana Carolina B B Candido
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
| | - Thaís A S Oliveira
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Alexandre Borges
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
- Faculty of Medicine, University Center of Santa Fe do Sul, Santa Fé do Sul 15775-000, SP, Brazil
| | - Nicoli D Oliveira
- Animal Science Post Graduation, University of Franca, Franca 14404-600, SP, Brazil
| | - Carla D Lopes
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Renato P Orenha
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
| | - Renato L T Parreira
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
| | - Antônio E M Crotti
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Lizandra G Magalhães
- Research Group on Natural Products, Center for Research in Sciences and Technology, University of Franca, Franca 14404-600, SP, Brazil
- Animal Science Post Graduation, University of Franca, Franca 14404-600, SP, Brazil
| |
Collapse
|
29
|
Impact of Laboratory-Adapted Intracellular Trypanosoma cruzi Strains on the Activity Profiles of Compounds with Anti- T. cruzi Activity. Microorganisms 2023; 11:microorganisms11020476. [PMID: 36838441 PMCID: PMC9967867 DOI: 10.3390/microorganisms11020476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 02/16/2023] Open
Abstract
Chagas disease is caused by infection with the protozoan parasite, Trypanosoma cruzi. The disease causes ~12,000 deaths annually and is one of the world's 20 neglected tropical diseases, as defined by the World Health Organisation. The drug discovery pipeline for Chagas disease currently has few new clinical candidates, with high attrition rates an ongoing issue. To determine if the Trypanosoma cruzi strain utilised to assess in vitro compound activity impacts activity, a comparison of laboratory-adapted T. cruzi strains from differing geographical locations was undertaken for a selection of compounds with anti-T. cruzi activity. To minimise the possible effect of differences in experimental methodology, the same host cell and multiplicity of infection were utilised. To determine whether the compound exposure time influenced results, activity was determined following exposure for 48 and 72 h of incubation. To ascertain whether replication rates affected outcomes, comparative rates of replication of the T. cruzi strains were investigated, using the nucleoside analogue, 5-ethynyl-2'-deoxyuridine. Minimal differences in the in vitro activity of compounds between strains were observed following 48 h incubation, whereas significant differences were observed following 72 h incubation, in particular for the cytochrome P450 inhibitors tested and the cell cycle inhibitor, camptothecin. Thus, the use of panels of laboratory adapted strains in vitro may be dependent on the speed of action that is prioritised. For the identification of fast-acting compounds, an initial shorter duration assay using a single strain may be used. A longer incubation to identify compound activity may alternatively require profiling of compounds against multiple T. cruzi strains.
Collapse
|
30
|
de Oliveira Rezende Júnior C, Martinez PDG, Ferreira RAA, Koovits PJ, Miranda Soares B, Ferreira LLG, Michelan-Duarte S, Chelucci RC, Andricopulo AD, Matheeussen A, Van Pelt N, Caljon G, Maes L, Campbell S, Kratz JM, Mowbray CE, Dias LC. Hit-to-lead optimization of a 2-aminobenzimidazole series as new candidates for chagas disease. Eur J Med Chem 2023; 246:114925. [PMID: 36459758 DOI: 10.1016/j.ejmech.2022.114925] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022]
Abstract
Chagas disease is a neglected tropical disease caused by Trypanosoma cruzi. Because current treatments present several limitations, including long duration, variable efficacy and serious side effects, there is an urgent need to explore new antitrypanosomal drugs. The present study describes the hit-to-lead optimization of a 2-aminobenzimidazole hit 1 identified through in vitro phenotypic screening of a chemical library against intracellular Trypanosoma cruzi amastigotes, which focused on optimizing potency, selectivity, microsomal stability and lipophilicity. Multiparametric Structure-Activity Relationships were investigated using a set of 277 derivatives. Although the physicochemical and biological properties of the initial hits were improved, a combination of low kinetic solubility and in vitro cytotoxicity against mammalian cells prevented progression of the best compounds to an efficacy study using a mouse model of Chagas disease.
Collapse
Affiliation(s)
| | | | | | - Paul John Koovits
- Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, 13083-861, Brazil
| | - Bruna Miranda Soares
- Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, 13083-861, Brazil
| | - Leonardo L G Ferreira
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of São Carlos, University of São Paulo (USP), São Carlos, SP, 13563-120, Brazil
| | - Simone Michelan-Duarte
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of São Carlos, University of São Paulo (USP), São Carlos, SP, 13563-120, Brazil
| | - Rafael Consolin Chelucci
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of São Carlos, University of São Paulo (USP), São Carlos, SP, 13563-120, Brazil
| | - Adriano D Andricopulo
- Laboratory of Medicinal and Computational Chemistry, Physics Institute of São Carlos, University of São Paulo (USP), São Carlos, SP, 13563-120, Brazil
| | - An Matheeussen
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Universiteitsplein 1, 2610, Antwerpen, Belgium
| | - Natascha Van Pelt
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Universiteitsplein 1, 2610, Antwerpen, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Universiteitsplein 1, 2610, Antwerpen, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Universiteitsplein 1, 2610, Antwerpen, Belgium
| | - Simon Campbell
- Drugs for Neglected Diseases Initiative (DNDi), 15 Chemin Camille-Vidart, 1202, Geneva, Switzerland
| | - Jadel M Kratz
- Drugs for Neglected Diseases Initiative (DNDi), 15 Chemin Camille-Vidart, 1202, Geneva, Switzerland
| | - Charles E Mowbray
- Drugs for Neglected Diseases Initiative (DNDi), 15 Chemin Camille-Vidart, 1202, Geneva, Switzerland
| | - Luiz Carlos Dias
- Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, 13083-861, Brazil.
| |
Collapse
|
31
|
García F, Musikant D, Escalona JL, Edreira MM, Liñares GG. Lipase-Catalyzed Synthesis and Biological Evaluation of N-Picolineamides as Trypanosoma cruzi Antiproliferative Agents. ACS Med Chem Lett 2023; 14:59-65. [PMID: 36655123 PMCID: PMC9841590 DOI: 10.1021/acsmedchemlett.2c00425] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
In our search for new safe antiparasitic agents, an enzymatic pathway was applied to synthesize a series of N-pyridinylmethyl amides derived from structurally different carboxylic acids. Thirty derivatives, including 11 new compounds, were prepared through lipase-catalyzed acylation in excellent yields. In order to optimize the synthetic methodology, the impact of different reaction parameters was analyzed. Some compounds were evaluated as antiproliferative agents against Trypanosoma cruzi, the parasite responsible for American trypanosomiasis (Chagas' disease). Some of them showed significant activity as parasite proliferation inhibitors. Amides derived from 2-aminopicoline and stearic and elaidic acids were as potent as nifurtimox against the amastigote form of T. cruzi, the clinically relevant form of the parasite. Even more, a powerful synergism between nifurtimox and N-(pyridin-2-ylmethyl)stereamide was observed, almost completely inhibiting the proliferation of the parasite. Besides, the obtained compounds showed no toxicity in Vero cells, making them excellent potential candidates as lead drugs.
Collapse
Affiliation(s)
- Fabricio
Freije García
- Laboratorio
de Biocatálisis, Departamento de Química Orgánica
y UMYMFOR, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET, Ciudad Universitaria, Pabellón 2, piso 3, C1428EGA Buenos Aires, Argentina
| | - Daniel Musikant
- Laboratorio
de Biología Molecular de Trypanosomas, Departamento de Química
Biológica e IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET, Ciudad Universitaria, Pabellón 2, piso 4, C1428EGA Buenos Aires, Argentina
| | - José L. Escalona
- Laboratorio
de Biología Molecular de Trypanosomas, Departamento de Química
Biológica e IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET, Ciudad Universitaria, Pabellón 2, piso 4, C1428EGA Buenos Aires, Argentina
| | - Martín M. Edreira
- Laboratorio
de Biología Molecular de Trypanosomas, Departamento de Química
Biológica e IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET, Ciudad Universitaria, Pabellón 2, piso 4, C1428EGA Buenos Aires, Argentina
| | - Guadalupe García Liñares
- Laboratorio
de Biocatálisis, Departamento de Química Orgánica
y UMYMFOR, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET, Ciudad Universitaria, Pabellón 2, piso 3, C1428EGA Buenos Aires, Argentina
| |
Collapse
|
32
|
Tawaraishi T, Ochida A, Akao Y, Itono S, Kamaura M, Akther T, Shimada M, Canan S, Chowdhury S, Cao Y, Condroski K, Engkvist O, Francisco A, Ghosh S, Kaki R, Kelly JM, Kimura C, Kogej T, Nagaoka K, Naito A, Pairaudeau G, Radu C, Roberts I, Shum D, Watanabe NA, Xie H, Yonezawa S, Yoshida O, Yoshida R, Mowbray C, Perry B. Collaborative Virtual Screening Identifies a 2-Aryl-4-aminoquinazoline Series with Efficacy in an In Vivo Model of Trypanosoma cruzi Infection. J Med Chem 2023; 66:1221-1238. [PMID: 36607408 PMCID: PMC9884087 DOI: 10.1021/acs.jmedchem.2c00775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Probing multiple proprietary pharmaceutical libraries in parallel via virtual screening allowed rapid expansion of the structure-activity relationship (SAR) around hit compounds with moderate efficacy against Trypanosoma cruzi, the causative agent of Chagas Disease. A potency-improving scaffold hop, followed by elaboration of the SAR via design guided by the output of the phenotypic virtual screening efforts, identified two promising hit compounds 54 and 85, which were profiled further in pharmacokinetic studies and in an in vivo model of T. cruzi infection. Compound 85 demonstrated clear reduction of parasitemia in the in vivo setting, confirming the interest in this series of 2-(pyridin-2-yl)quinazolines as potential anti-trypanosome treatments.
Collapse
Affiliation(s)
- Taisuke Tawaraishi
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Atsuko Ochida
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yuichiro Akao
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Sachiko Itono
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masahiro Kamaura
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Thamina Akther
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Mitsuyuki Shimada
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Stacie Canan
- Celgene
Corporation, Celgene Global Health, 10300 Campus Point Drive, San Diego, California 92121, United States
| | - Sanjoy Chowdhury
- TCG
Lifesciences, Plot No-7,
Salt Lake Electronics Complex, BN Block, Sector V, Kolkata 700091, India
| | - Yafeng Cao
- WuXi
AppTec Company Ltd., 666 Gaoxin Road, East Lake High-Tech Development Zone, Wuhan 430075, People’s Republic of China
| | - Kevin Condroski
- Celgene
Corporation, Celgene Global Health, 10300 Campus Point Drive, San Diego, California 92121, United States
| | - Ola Engkvist
- AstraZeneca
Discovery Sciences, R&D, Pepparedsleden 1, 431 50 Mölndal, Sweden
| | - Amanda Francisco
- London School
of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K.
| | - Sunil Ghosh
- TCG
Lifesciences, Plot No-7,
Salt Lake Electronics Complex, BN Block, Sector V, Kolkata 700091, India
| | - Rina Kaki
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - John M. Kelly
- London School
of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K.
| | - Chiaki Kimura
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Thierry Kogej
- AstraZeneca
Discovery Sciences, R&D, Pepparedsleden 1, 431 50 Mölndal, Sweden
| | - Kazuya Nagaoka
- Eisai
Co., Ltd, 1-3, Tokodai
5-chome, Tsukuba, Ibaraki 300-2635, Japan
| | - Akira Naito
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Garry Pairaudeau
- AstraZeneca,
Discovery Sciences, R&D, The Darwin Building, 310 Milton Road, Milton, Cambridge CB4 0WG, U.K.
| | - Constantin Radu
- Institut
Pasteur Korea, 16, Daewangpangyo-ro
712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Ieuan Roberts
- AstraZeneca,
Discovery Sciences, R&D, The Darwin Building, 310 Milton Road, Milton, Cambridge CB4 0WG, U.K.
| | - David Shum
- Institut
Pasteur Korea, 16, Daewangpangyo-ro
712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Nao-aki Watanabe
- Eisai
Co., Ltd, 1-3, Tokodai
5-chome, Tsukuba, Ibaraki 300-2635, Japan
| | - Huanxu Xie
- WuXi
AppTec Company Ltd., 666 Gaoxin Road, East Lake High-Tech Development Zone, Wuhan 430075, People’s Republic of China
| | - Shuji Yonezawa
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Osamu Yoshida
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Ryu Yoshida
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Charles Mowbray
- Drugs for Neglected
Diseases Initiative, 15 Chemin Camille Vidart, Geneva 1202, Switzerland
| | - Benjamin Perry
- Drugs for Neglected
Diseases Initiative, 15 Chemin Camille Vidart, Geneva 1202, Switzerland,
| |
Collapse
|
33
|
Identification of Aryl Polyamines Derivatives as Anti- Trypanosoma cruzi Agents Targeting Iron Superoxide Dismutase. Pharmaceutics 2022; 15:pharmaceutics15010140. [PMID: 36678771 PMCID: PMC9863987 DOI: 10.3390/pharmaceutics15010140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/13/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023] Open
Abstract
Chagas disease (CD) is a tropical and potentially fatal infection caused by Trypanosoma cruzi. Although CD was limited to Latin America as a silent disease, CD has become widespread as a result of globalization. Currently, 6-8 million people are infected worldwide, and no effective treatment is available. Here, we identify new effective agents against T. cruzi. In short, 16 aryl polyamines were screened in vitro against different T. cruzi strains, and lead compounds were evaluated in vivo after oral administration in both the acute and chronic infections. The mode of action was also evaluated at the energetic level, and its high activity profile could be ascribed to a mitochondria-dependent bioenergetic collapse and redox stress by inhibition of the Fe-SOD enzyme. We present compound 15 as a potential compound that provides a step forward for the development of new agents to combat CD.
Collapse
|
34
|
Osorio-Nieto U, Salas CO, Mendez-Alvarez D, Rivera G, Moreno-Rodriguez A, Perez-Cervera Y, Castillo-Real LM, Espinosa-Bustos C. 2,3-Diketopiperazine as potential scaffold to develop new anti-Chagasic agents. Med Chem Res 2022. [DOI: 10.1007/s00044-022-03003-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
35
|
Katchborian-Neto A, Santos MFC, Vilas-Boas DF, Dos Santos EG, Veloso MP, Bueno PCP, Caldas IS, Soares MG, Dias DF, Chagas-Paula DA. Immunological Modulation and Control of Parasitaemia by Ayahuasca Compounds: Therapeutic Potential for Chagas's Disease. Chem Biodivers 2022; 19:e202200409. [PMID: 36163588 DOI: 10.1002/cbdv.202200409] [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: 04/28/2022] [Accepted: 09/08/2022] [Indexed: 11/12/2022]
Abstract
Ayahuasca is a psychoactive and psychedelic decoct composed mainly of Banisteriopsis caapi and Psychotria viridis plant species. The beverage is rich in alkaloids and it is ritualistically used by several indigenous communities of South America as a natural medicine. There are also reports in the literature indicating the prophylaxis potential of Ayahuasca alkaloids against internal parasites. In the present study, Ayahuasca exhibited moderate in vitro activity against Trypanosoma cruzi trypomastigotes (IC50 95.78 μg/mL) compared to the reference drug benznidazole (IC50 2.03 μg/mL). The β-carboline alkaloid harmine (HRE), isolated from B. caapi, was considered active against the trypomastigotes forms (IC50 6.37), and the tryptamine N, N-dimethyltryptamine (DMT), isolated from P. viridis was also moderately active with IC50 of 21.02 μg/mL. Regarding the in vivo evaluations, no collateral effects were observed. The HRE alone demonstrated the highest trypanocidal activity in a dose-responsive manner (10 and 100 mg/kg). The Ayahuasca and the association between HRE and DMT worsened the parasitaemia, suggesting a modulation of the immunological response during the T. cruzi infection, especially by increasing total Immunoglobulin (IgG) and IgG1 antibody levels. The in silico molecular docking revealed HRE binding with low energy at two sites of the Trypanothione reductase enzyme (TR), which are absent in humans, and thus considered a promissory target for drug discovery. In conclusion, Ayahuasca compounds seem to not be toxic at the concentrations of the in vivo evaluations and can promote trypanocidal effect in multi targets, including control of parasitaemia, immunological modulation and TR enzymatic inhibition, which might benefit the treatments of patients with Chagas' disease. Moreover, the present study also provides scientific information to support the prophylactic potential of Ayahuasca against internal parasites.
Collapse
Affiliation(s)
- Albert Katchborian-Neto
- Institute of Chemistry, Federal University of Alfenas, Rua Gabriel Monteiro da Silva 700, 37130-001, Alfenas, Minas Gerais, Brazil
| | - Mário Ferreira Conceição Santos
- Department of Chemistry and Physics, Center of Exact, Natural and Health Sciences, Federal University of Espírito Santo, Alto Universitário, 29500-000, Alegre, Espírito Santo, Brazil
| | - Diego Fernandes Vilas-Boas
- Institute of Biomedical Sciences, Department of Pathology and Basic Parasitology, Federal University of Alfenas, Rua Gabriel Monteiro da Silva 700, Alfenas, Minas Gerais, 37131-000, Brazil
| | - Elda Gonçalves Dos Santos
- Institute of Biomedical Sciences, Department of Pathology and Basic Parasitology, Federal University of Alfenas, Rua Gabriel Monteiro da Silva 700, Alfenas, Minas Gerais, 37131-000, Brazil
| | - Márcia Paranho Veloso
- Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Rua Gabriel Monteiro da Silva, 700, Alfenas, Minas Gerais, 37130-000, Brazil
| | - Paula Carolina Pires Bueno
- Institute of Chemistry, Federal University of Alfenas, Rua Gabriel Monteiro da Silva 700, 37130-001, Alfenas, Minas Gerais, Brazil
| | - Ivo Santana Caldas
- Institute of Biomedical Sciences, Department of Pathology and Basic Parasitology, Federal University of Alfenas, Rua Gabriel Monteiro da Silva 700, Alfenas, Minas Gerais, 37131-000, Brazil
| | - Marisi Gomes Soares
- Institute of Chemistry, Federal University of Alfenas, Rua Gabriel Monteiro da Silva 700, 37130-001, Alfenas, Minas Gerais, Brazil
| | - Danielle Ferreira Dias
- Institute of Chemistry, Federal University of Alfenas, Rua Gabriel Monteiro da Silva 700, 37130-001, Alfenas, Minas Gerais, Brazil
| | - Daniela Aparecida Chagas-Paula
- Institute of Chemistry, Federal University of Alfenas, Rua Gabriel Monteiro da Silva 700, 37130-001, Alfenas, Minas Gerais, Brazil
| |
Collapse
|
36
|
Ang CW, Lee BM, Jackson CJ, Wang Y, Franzblau SG, Francisco AF, Kelly JM, Bernhardt PV, Tan L, West NP, Sykes ML, Hinton AO, Bolisetti R, Avery VM, Cooper MA, Blaskovich MA. Nitroimidazopyrazinones with Oral Activity against Tuberculosis and Chagas Disease in Mouse Models of Infection. J Med Chem 2022; 65:13125-13142. [DOI: 10.1021/acs.jmedchem.2c00972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Chee Wei Ang
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
- School of Science, Monash University Malaysia, Subang Jaya, 47500 Selangor, Malaysia
| | - Brendon M. Lee
- Research School of Chemistry, Australian National University, Sullivans Creek Road, Acton ACT 2601, Australia
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medical College, New York, New York 10021, United States
| | - Colin J. Jackson
- Research School of Chemistry, Australian National University, Sullivans Creek Road, Acton ACT 2601, Australia
| | - Yuehong Wang
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Scott G. Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Amanda F. Francisco
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Paul V. Bernhardt
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Lendl Tan
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas P. West
- School of Chemistry and Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Melissa L. Sykes
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
| | - Alexandra O. Hinton
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Raghu Bolisetti
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Vicky M. Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
- School of Environment and Science, Griffith University, Nathan, Queensland 4111, Australia
| | - Matthew A. Cooper
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Mark A.T. Blaskovich
- Center for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
37
|
Martín-Escolano R, Rosales MJ, Marín C. Biological characteristics of the Trypanosoma cruzi Arequipa strain make it a good model for Chagas disease drug discovery. Acta Trop 2022; 236:106679. [PMID: 36096184 DOI: 10.1016/j.actatropica.2022.106679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/04/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022]
Abstract
Trypanosoma cruzi, the causative agent of Chagas disease (CD), is a genuine parasite with tremendous genetic diversity and a complex life cycle. Scientists have studied this disease for more than 100 years, and CD drug discovery has been a mainstay due to the absence of an effective treatment. Technical advances in several areas have contributed to a better understanding of the complex biology and life cycle of this parasite, with the aim of designing the ideal profile of both drug and therapeutic options to treat CD. Here, we present the T. cruzi Arequipa strain (MHOM/Pe/2011/Arequipa) as an interesting model for CD drug discovery. We characterized acute-phase parasitaemia and chronic-phase tropism in BALB/c mice and determined the in vitro and in vivo benznidazole susceptibility profile of the different morphological forms of this strain. The tropism of this strain makes it an interesting model for the screening of new compounds with a potential anti-Chagas profile for the treatment of this disease.
Collapse
Affiliation(s)
- Rubén Martín-Escolano
- Laboratory of Molecular & Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK.
| | - María José Rosales
- Department of Parasitology, University of Granada, Severo Ochoa s/n, Granada 18071, Spain
| | - Clotilde Marín
- Department of Parasitology, University of Granada, Severo Ochoa s/n, Granada 18071, Spain.
| |
Collapse
|
38
|
Barbosa JMC, Pedra-Rezende Y, Pereira LD, de Melo TG, Barbosa HS, Lannes-Vieira J, de Castro SL, Daliry A, Salomão K. Benznidazole and amiodarone combined treatment attenuates cytoskeletal damage in Trypanosoma cruzi-infected cardiac cells. Front Cell Infect Microbiol 2022; 12:975931. [PMID: 36093188 PMCID: PMC9452897 DOI: 10.3389/fcimb.2022.975931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/05/2022] [Indexed: 12/02/2022] Open
Abstract
Chagas disease (CD), a neglected tropical disease caused by the protozoan parasite Trypanosoma cruzi, is an important public health problem mainly in Latin America, leading to approximately 12,000 annual deaths. Current etiological treatment for CD is limited to two nitro compounds, benznidazole (Bz) and nifurtimox (Nif), both presenting relevant limitations. Different approaches have been employed to establish more effective and safer schemes to treat T. cruzi infection, mostly based on drug repurposing and combination therapies. Amiodarone (AMD), an antiarrhythmic medicament of choice for patients with the chronic cardiac form of CD, is also recognized as a trypanocidal agent. Therefore, our aim is to investigate the combined treatment Bz + AMD on trypomastigote viability, control of T. cruzi intracellular form proliferation, and recovery of the infection-induced cytoskeleton alterations in cardiac cells. The combination of Bz + AMD did not improve the direct trypanocidal effect of AMD on the infective blood trypomastigote and replicative intracellular forms of the parasite. Otherwise, the treatment of T. cruzi-infected cardiac cells with Bz plus AMD attenuated the infection-triggered cytoskeleton damage of host cells and the cytotoxic effects of AMD. Thus, the combined treatment Bz + AMD may favor parasite control and hamper tissue damage.
Collapse
Affiliation(s)
| | | | | | | | - Helene Santos Barbosa
- Laboratóriode de Biologia Estrutural, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Anissa Daliry
- Laboratório de Investigação Cardiovascular, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Kelly Salomão
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
- *Correspondence: Kelly Salomão,
| |
Collapse
|
39
|
Martinho ACC, Resende DDM, Landin ES, Dit Lapierre TJWJ, Bernardes TCD, Martins LC, Ferreira RS, Murta SMF, de Oliveira Rezende Júnior C. Synthesis, Design, and Structure-Activity Relationship of a Benzenesulfonylpiperazine Series Against Trypanosoma Cruzi. ChemMedChem 2022; 17:e202200211. [PMID: 35993440 DOI: 10.1002/cmdc.202200211] [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: 04/13/2022] [Revised: 08/19/2022] [Indexed: 11/09/2022]
Abstract
Chagas disease is a neglected tropical disease, endemic in Latin America and caused by the protozoan parasite Trypanosoma cruzi . Available treatments show low cure efficacy during the chronic phase of the disease and cause a series of side effects, reinforcing the need to develop new drugs against Chagas disease. In this work, we describe the optimization of a trypanocidal hit compound recently reported in phenotypic HTS studies against Trypanosoma cruzi . A hit-to-lead process was initiated and a structure-activity relationship against Trypanosoma cruzi was obtained after the synthesis and biological evaluation of 22 new benzenesulfonylpiperazine derivatives. From this SAR study, we identified three compounds with a promising predicted ADMET profile and potency comparable to the reference drug benznidazole, which are candidates for further development towards therapies for Chagas disease.
Collapse
Affiliation(s)
| | - Daniela de Melo Resende
- Fundação Oswaldo Cruz Instituto René Rachou: Fundacao Oswaldo Cruz Instituto Rene Rachou, René Rachou Institute, BRAZIL
| | - Emanuelly Silva Landin
- Federal University of Uberlandia: Universidade Federal de Uberlandia, Chemistry Institute, BRAZIL
| | | | | | - Luan Carvalho Martins
- Federal University of Minas Gerais: Universidade Federal de Minas Gerais, Biochemistry and immunology, BRAZIL
| | - Rafaela Salgado Ferreira
- Federal University of Minas Gerais: Universidade Federal de Minas Gerais, Biochemistry and immunology, BRAZIL
| | - Silvane Maria Fonseca Murta
- Oswaldo Cruz Foundation Rene Rachou Institute: Fundacao Oswaldo Cruz Instituto Rene Rachou, René Rachou Institute, BRAZIL
| | | |
Collapse
|
40
|
McGonagle K, Tarver GJ, Cantizani J, Cotillo I, Dodd PG, Ferguson L, Gilbert IH, Marco M, Miles T, Naylor C, Osuna-Cabello M, Paterson C, Read KD, Pinto EG, Riley J, Scullion P, Shishikura Y, Simeons F, Stojanovski L, Svensen N, Thomas J, Wyatt PG, Manzano P, De Rycker M, Thomas MG. Identification and development of a series of disubstituted piperazines for the treatment of Chagas disease. Eur J Med Chem 2022; 238:114421. [PMID: 35594652 PMCID: PMC11458808 DOI: 10.1016/j.ejmech.2022.114421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 11/23/2022]
Abstract
Approximately 6-7 million people around the world are estimated to be infected with Trypanosoma cruzi, the causative agent of Chagas disease. The current treatments are inadequate and therefore new medical interventions are urgently needed. In this paper we describe the identification of a series of disubstituted piperazines which shows good potency against the target parasite but is hampered by poor metabolic stability. We outline the strategies used to mitigate this issue such as lowering logD, bioisosteric replacements of the metabolically labile piperazine ring and use of plate-based arrays for quick diversity scoping. We discuss the success of these strategies within the context of this series and highlight the challenges faced in phenotypic programs when attempting to improve the pharmacokinetic profile of compounds whilst maintaining potency against the desired target.
Collapse
Affiliation(s)
- Kate McGonagle
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Gary J Tarver
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Juan Cantizani
- Global Health R&D, GlaxoSmithKline, Tres Cantos, 28760, Spain
| | - Ignacio Cotillo
- Global Health R&D, GlaxoSmithKline, Tres Cantos, 28760, Spain
| | - Peter G Dodd
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Liam Ferguson
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Ian H Gilbert
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Maria Marco
- Global Health R&D, GlaxoSmithKline, Tres Cantos, 28760, Spain
| | - Tim Miles
- Global Health R&D, GlaxoSmithKline, Tres Cantos, 28760, Spain
| | - Claire Naylor
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Maria Osuna-Cabello
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Christy Paterson
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Kevin D Read
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Erika G Pinto
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Jennifer Riley
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Paul Scullion
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Yoko Shishikura
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Frederick Simeons
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Laste Stojanovski
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Nina Svensen
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - John Thomas
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Paul G Wyatt
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK
| | - Pilar Manzano
- Global Health R&D, GlaxoSmithKline, Tres Cantos, 28760, Spain.
| | - Manu De Rycker
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK.
| | - Michael G Thomas
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, UK.
| |
Collapse
|
41
|
5-Nitroindazole-based compounds: further studies for activity optimization as anti-Trypanosoma cruzi agents. Acta Trop 2022; 234:106607. [PMID: 35907502 DOI: 10.1016/j.actatropica.2022.106607] [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: 05/19/2022] [Revised: 06/19/2022] [Accepted: 07/12/2022] [Indexed: 11/20/2022]
Abstract
In this study, a new series of eleven 5-nitroindazole derivatives (10-20) and a related 6-nitroquinazoline (21) was synthesized and tested in vitro against different forms of the kinetoplastid parasite Trypanosoma cruzi, etiological agent of Chagas disease. Among these compounds, derivatives 11-14 and 17 showed trypanocidal profiles on epimastigotes (IC50 = 1.00-8.75 µM) considerably better than that of the reference drug benznidazole, BZ (IC50 = 25.22 µM). Furthermore, the lack of cytotoxicity observed for compounds 11, 12, 14, 17 and 18 over L929 fibroblasts, led to a notable selectivity (SI) on the extracellular replicative form of the parasite: SIEPI > 12.41 to > 256 µM. Since these five derivatives overpassed the cut-off value established by BZ (SIEPI ≥ 10), they were moved to a more specific assay against the intracellular and replicative form of T. cruzi, i.e, amastigotes. These molecules were not as active as BZ (IC50 = 0.57 µM) against this parasite form; however, all of them showed remarkable IC50 values lower than 7 µM. Special mention deserve compounds 12 and 17, whose SIAMA were > 246.15 and > 188.23, respectively. The results compiled in the present work, point to a positive impact over the trypanocidal activity of the electron withdrawing substituents introduced at position 2 of the N-2 benzyl moiety of these compounds, especially fluorine, i.e., derivatives 12 and 17. These outcomes, supported by the in silico prediction of good oral bioavailability and suitable risk profile, reinforce the 5-nitroindazole scaffold as an adequate template for preparing potential antichagasic agents.
Collapse
|
42
|
Temporal and Wash-Out Studies Identify Medicines for Malaria Venture Pathogen Box Compounds with Fast-Acting Activity against Both Trypanosoma cruzi and Trypanosoma brucei. Microorganisms 2022; 10:microorganisms10071287. [PMID: 35889006 PMCID: PMC9317670 DOI: 10.3390/microorganisms10071287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Chagas disease caused by the protozoan Trypanosoma cruzi is endemic to 21 countries in the Americas, effects approximately 6 million people and on average results in 12,000 deaths annually. Human African Trypanosomiasis (HAT) is caused by the Trypanosoma brucei sub-species, endemic to 36 countries within sub-Saharan Africa. Treatment regimens for these parasitic diseases are complicated and not effective against all disease stages; thus, there is a need to find improved treatments. To identify new molecules for the drug discovery pipelines for these diseases, we have utilised in vitro assays to identify compounds with selective activity against both T. cruzi and T.b. brucei from the Medicines for Malaria Venture (MMV) Pathogen Box compound collection. To prioritise these molecules for further investigation, temporal and wash off assays were utilised to identify the speed of action and cidality of compounds. For translational relevance, compounds were tested against clinically relevant T.b. brucei subspecies. Compounds with activity against T. cruzi cytochrome P450 (TcCYP51) have not previously been successful in clinical trials for chronic Chagas disease; thus, to deprioritise compounds with this activity, they were tested against recombinant TcCYP51. Compounds with biological profiles warranting progression offer important tools for drug and target development against kinetoplastids.
Collapse
|
43
|
Martín-Escolano J, Marín C, Rosales MJ, Tsaousis AD, Medina-Carmona E, Martín-Escolano R. An Updated View of the Trypanosoma cruzi Life Cycle: Intervention Points for an Effective Treatment. ACS Infect Dis 2022; 8:1107-1115. [PMID: 35652513 PMCID: PMC9194904 DOI: 10.1021/acsinfecdis.2c00123] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Chagas disease (CD)
is a parasitic, systemic, chronic, and often
fatal illness caused by infection with the protozoan Trypanosoma
cruzi. The World Health Organization classifies CD as the
most prevalent of poverty-promoting neglected tropical diseases, the
most important parasitic one, and the third most infectious disease
in Latin America. Currently, CD is a global public health issue that
affects 6–8 million people. However, the current approved treatments
are limited to two nitroheterocyclic drugs developed more than 50
years ago. Many efforts have been made in recent decades to find new
therapies, but our limited understanding of the infection process,
pathology development, and long-term nature of this disease has made
it impossible to develop new drugs, effective treatment, or vaccines.
This Review aims to provide a comprehensive update on our understanding
of the current life cycle, new morphological forms, and genetic diversity
of T. cruzi, as well as identify intervention points
in the life cycle where new drugs and treatments could achieve a parasitic
cure.
Collapse
Affiliation(s)
- Javier Martín-Escolano
- Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, E41013 Seville, Spain
| | - Clotilde Marín
- Department of Parasitology, University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | - María J. Rosales
- Department of Parasitology, University of Granada, Severo Ochoa s/n, 18071 Granada, Spain
| | - Anastasios D. Tsaousis
- Laboratory of Molecular & Evolutionary Parasitology, RAPID group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, U.K
| | - Encarnación Medina-Carmona
- Department of Physical Chemistry, University of Granada, 18071 Granada, Spain
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, U.K
| | - Rubén Martín-Escolano
- Laboratory of Molecular & Evolutionary Parasitology, RAPID group, School of Biosciences, University of Kent, Canterbury CT2 7NJ, U.K
| |
Collapse
|
44
|
Kratz JM, Gonçalves KR, Romera LM, Moraes CB, Bittencourt-Cunha P, Schenkman S, Chatelain E, Sosa-Estani S. The translational challenge in Chagas disease drug development. Mem Inst Oswaldo Cruz 2022; 117:e200501. [PMID: 35613156 PMCID: PMC9128742 DOI: 10.1590/0074-02760200501] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
Chagas disease is a neglected tropical disease caused by the protozoan parasite Trypanosoma cruzi. There is an urgent need for safe, effective, and accessible new treatments since the currently approved drugs have serious limitations. Drug development for Chagas disease has historically been hampered by the complexity of the disease, critical knowledge gaps, and lack of coordinated R&D efforts. This review covers some of the translational challenges associated with the progression of new chemical entities from preclinical to clinical phases of development, and discusses how recent technological advances might allow the research community to answer key questions relevant to the disease and to overcome hurdles in R&D for Chagas disease.
Collapse
Affiliation(s)
- Jadel M Kratz
- Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Karolina R Gonçalves
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brasil
| | - Lavínia Md Romera
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brasil
| | - Carolina Borsoi Moraes
- Universidade Federal de São Paulo, Departamento de Ciências Farmacêuticas, Diadema, SP, Brasil
| | - Paula Bittencourt-Cunha
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brasil.,Universidade Federal de São Paulo, Departamento de Microbiologia, Imunologia e Parasitologia, São Paulo, SP, Brasil
| | - Sergio Schenkman
- Universidade Federal de São Paulo, Departamento de Microbiologia, Imunologia e Parasitologia, São Paulo, SP, Brasil
| | - Eric Chatelain
- Drugs for Neglected Diseases initiative, Geneva, Switzerland
| | - Sergio Sosa-Estani
- Drugs for Neglected Diseases initiative, Geneva, Switzerland.,Epidemiology and Public Health Research Centre, CIESP-CONICET, Buenos Aires, Argentina
| |
Collapse
|
45
|
Synthesis and study of the trypanocidal activity of catechol-containing 3-arylcoumarins, inclusion in β-cyclodextrin complexes and combination with benznidazole. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2021.103641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
46
|
Experimental Combination Therapy with Amiodarone and Low-Dose Benznidazole in a Mouse Model of Trypanosoma cruzi Acute Infection. Microbiol Spectr 2022; 10:e0185221. [PMID: 35138142 PMCID: PMC8826820 DOI: 10.1128/spectrum.01852-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Chagas disease (CD), caused by Trypanosoma cruzi, affects approximately 6 to 7 million people in Latin America, with cardiomyopathy being the clinical manifestation most commonly associated with patient death during the acute phase. The etiological treatment of CD is restricted to benznidazole (Bz) and nifurtimox (Nif), which involve long periods of administration, frequent side effects, and low efficacy in the chronic phase. Thus, combined therapies emerge as an important tool in the treatment of CD, allowing the reduction of Bz dose and treatment duration. In this sense, amiodarone (AMD), the most efficient antiarrhythmic drug currently available and prescribed to CD patients, is a potential candidate for combined treatment due to its known trypanocidal activity. However, the efficacy of AMD during the acute phase of CD and its interaction with Bz or Nif are still unknown. In the present study, using a well-established murine model of the acute phase of CD, we observed that the Bz/AMD combination was more effective in reducing the peak parasitemia than both monotherapy treatments. Additionally, the Bz/AMD combination reduced (i) interleukin-6 (IL-6) levels in cardiac tissue, (ii) P-wave duration, and (iii) frequency of arrhythmia in infected animals and (iv) restored gap junction integrity in cardiac tissue. Therefore, our study validates AMD as a promising candidate for combined therapy with Bz, reinforcing the strategy of combined therapy for CD. IMPORTANCE Chagas disease affects approximately 6 to 7 million people worldwide, with cardiomyopathy being the clinical manifestation that most commonly leads to patient death. The etiological treatment of Chagas disease is limited to drugs (benznidazole and nifurtimox) with relatively high toxicity and therapeutic failures. In this sense, amiodarone, the most effective currently available antiarrhythmic drug prescribed to patients with Chagas disease, is a potential candidate for combined treatment due to its known trypanocidal effect. In the present study, we show that combined treatment with benznidazole and amiodarone improves the trypanocidal effect and reduces cardiac damage in acutely T. cruzi-infected mice.
Collapse
|
47
|
Lascano F, García Bournissen F, Altcheh J. Review of pharmacological options for the treatment of Chagas disease. Br J Clin Pharmacol 2022; 88:383-402. [PMID: 33314266 DOI: 10.1111/bcp.14700] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/09/2020] [Accepted: 12/02/2020] [Indexed: 12/20/2022] Open
Abstract
Chagas disease (CD) is a worldwide problem, with over 8 million people infected in both rural and urban areas. CD was first described over a century ago, but only two drugs are currently available for CD treatment: benznidazole (BZN) and nifurtimox (NF). Treating CD-infected patients, especially children and women of reproductive age, is vital in order to prevent long-term sequelae, such as heart and gastrointestinal dysfunction, but this aim is still far from being accomplished. Currently, the strongest data to support benefit-risk considerations come from trials in children. Treatment response biomarkers need further development as serology is being questioned as the best method to assess treatment response. This article is a narrative review on the pharmacology of drugs for CD, particularly BZN and NF. Data on drug biopharmaceutical characteristics, safety and efficacy of both drugs are summarized from a clinical perspective. Current data on alternative compounds under evaluation for CD treatment, and new possible treatment response biomarkers are also discussed. Early diagnosis and treatment of CD, especially in paediatric patients, is vital for an effective and safe use of the available drugs (i.e. BZN and NF). New biomarkers for CD are urgently needed for the diagnosis and evaluation of treatment efficacy, and to guide efforts from academia and pharmaceutical companies to accelerate the process of new drug development.
Collapse
Affiliation(s)
- Fernanda Lascano
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Gobierno de la Ciudad de la Nación Argentina, Buenos Aires, Argentina.,Servicio de Parasitología y Chagas, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Facundo García Bournissen
- Division of Pediatric Clinical Pharmacology, Department of Pediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada
| | - Jaime Altcheh
- Instituto Multidisciplinario de Investigaciones en Patologías Pediátricas (IMIPP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Gobierno de la Ciudad de la Nación Argentina, Buenos Aires, Argentina.,Servicio de Parasitología y Chagas, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| |
Collapse
|
48
|
Monroy MC, Penados D, Pineda J, Ruiz EL, Agreda EO, Alcantara B, Rodas A, Lange K, Weinberg D, Bazzani R, Marchiol A, Herazo R, Agrelo RS, Abril M, Chuit R. A multidisciplinary, collaborative, inter-agency and comprehensive approach for the control of Chagas Disease as a public health problem in Guatemala. Acta Trop 2022; 225:106157. [PMID: 34634265 DOI: 10.1016/j.actatropica.2021.106157] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/10/2021] [Accepted: 09/20/2021] [Indexed: 11/26/2022]
Abstract
The Pan American Health Organization (PAHO) has defined Chagas Disease hotspots in Central America associated with the vector Triatoma spp. Triatoma dimidiata is a native vector adapted to multiple environments, including intra-domestic and peri-domestic habitats. A multi-institutional project named "Alliances for the elimination of Chagas in Central America" was created to help reduce the incidence of the disease in the region. Activities performed in the field as part of the project included aspects of vector surveillance and control, improvement of houses, diagnosis and treatment of individuals, health promotion, training of human resources and identification of access barriers to diagnosis and treatment. As a base line study, eleven villages, comprised of 1,572 households, were entomologically evaluated (83.4% overall participation); five were found to have very high infestation rates (>20%), three had high infestation rates (8-20%) and three had low-infestation rates (<8%), coinciding with the category of infestation-risk of the houses within each village. Serological tests were carried out in 812 people (>80% participation) in two of the 11 villages and none of the 128 children tested, less than 5 years of age, were positive for Trypanosoma cruzi infection. Community participation in all the activities was high (>70%). The collaboration between several subnational, national, and international institutions, each with specific roles, promoted community participation in the activities of vector control and patient care, thus, establishing a baseline to continue implementing and monitoring project progress.
Collapse
|
49
|
Assessing risk of vector transmission of Chagas disease through blood source analysis using LC-MS/MS for hemoglobin sequence identification. PLoS One 2022; 17:e0262552. [PMID: 35073364 PMCID: PMC8786159 DOI: 10.1371/journal.pone.0262552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/28/2021] [Indexed: 11/19/2022] Open
Abstract
Chagas disease is mainly transmitted by triatomine insect vectors that feed on vertebrate blood. The disease has complex domiciliary infestation patterns and parasite transmission dynamics, influenced by biological, ecological, and socioeconomic factors. In this context, feeding patterns have been used to understand vector movement and transmission risk. Recently, a new technique using Liquid chromatography tandem mass spectrometry (LC-MS/MS) targeting hemoglobin peptides has showed excellent results for understanding triatomines' feeding patterns. The aim of this study was to further develop the automated computational analysis pipeline for peptide sequence taxonomic identification, enhancing the ability to analyze large datasets data. We then used the enhanced pipeline to evaluate the feeding patterns of Triatoma dimidiata, along with domiciliary infestation risk variables, such as unkempt piles of firewood or construction material, cracks in bajareque and adobe walls and intradomiciliary animals. Our new python scripts were able to detect blood meal sources in 100% of the bugs analyzed and identified nine different species of blood meal sources. Human, chicken, and dog were the main blood sources found in 78.7%, 50.4% and 44.8% of the bugs, respectively. In addition, 14% of the bugs feeding on chicken and 15% of those feeding on dogs were captured in houses with no evidence of those animals being present. This suggests a high mobility among ecotopes and houses. Two of the three main blood sources, dog and chicken, were significantly (p < 0.05) affected by domiciliary infestation risk variables, including cracks in walls, construction material and birds sleeping in the intradomicile. This suggests that these variables are important for maintaining reproducing Triatoma dimidiata populations and that it is critical to mitigate these variables in all the houses of a village for effective control of these mobile vectors.
Collapse
|
50
|
Duchowicz PR, Fioressi SE, Bacelo DE. QSAR predictions on antichagas fenarimols. RESULTS IN CHEMISTRY 2022. [DOI: 10.1016/j.rechem.2021.100256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|