1
|
de Souza TP, Orlando LMR, Lara LDS, Paes VB, Dutra LP, dos Santos MS, Pereira MCDS. Synthesis and Anti- Trypanosoma cruzi Activity of New Pyrazole-Thiadiazole Scaffolds. Molecules 2024; 29:3544. [PMID: 39124949 PMCID: PMC11314410 DOI: 10.3390/molecules29153544] [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: 06/21/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Chagas disease, a silent but widespread disease that mainly affects a socioeconomically vulnerable population, lacks innovative safe drug therapy. The available drugs, benznidazole and nifurtimox, are more than fifty years old, have limited efficacy, and carry harmful side effects, highlighting the need for new therapeutics. This study presents two new series of pyrazole-thiadiazole compounds evaluated for trypanocidal activity using cellular models predictive of efficacy. Derivatives 1c (2,4-diCl) and 2k (4-NO2) were the most active against intracellular amastigotes. Derivative 1c also showed activity against trypomastigotes, with the detachment of the flagellum from the parasite body being a predominant effect at the ultrastructural level. Analogs have favorable physicochemical parameters and are predicted to be orally available. Drug efficacy was also evaluated in 3D cardiac microtissue, an important target tissue of Trypanosoma cruzi, with derivative 2k showing potent antiparasitic activity and a significant reduction in parasite load. Although 2k potentially reduced parasite load in the washout assay, it did not prevent parasite recrudescence. Drug combination analysis revealed an additive profile, which may lead to favorable clinical outcomes. Our data demonstrate the antiparasitic activity of pyrazole-thiadiazole derivatives and support the development of these compounds using new optimization strategies.
Collapse
Affiliation(s)
- Thamyris Perez de Souza
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz. Av. Brasil 4365, Rio de Janeiro 21040-900, RJ, Brazil; (T.P.d.S.); (L.M.R.O.); (L.d.S.L.); (V.B.P.)
| | - Lorraine Martins Rocha Orlando
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz. Av. Brasil 4365, Rio de Janeiro 21040-900, RJ, Brazil; (T.P.d.S.); (L.M.R.O.); (L.d.S.L.); (V.B.P.)
| | - Leonardo da Silva Lara
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz. Av. Brasil 4365, Rio de Janeiro 21040-900, RJ, Brazil; (T.P.d.S.); (L.M.R.O.); (L.d.S.L.); (V.B.P.)
| | - Vitoria Barbosa Paes
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz. Av. Brasil 4365, Rio de Janeiro 21040-900, RJ, Brazil; (T.P.d.S.); (L.M.R.O.); (L.d.S.L.); (V.B.P.)
| | - Lucas Penha Dutra
- Laboratório de Síntese de Sistemas Heterocíclicos (LaSSH), Instituto de Física e Química (IFQ), Universidade Federal de Itajubá, Av. BPS 1303, Pinheirinho, Itajubá 37500-903, MG, Brazil; (L.P.D.); (M.S.d.S.)
| | - Mauricio Silva dos Santos
- Laboratório de Síntese de Sistemas Heterocíclicos (LaSSH), Instituto de Física e Química (IFQ), Universidade Federal de Itajubá, Av. BPS 1303, Pinheirinho, Itajubá 37500-903, MG, Brazil; (L.P.D.); (M.S.d.S.)
| | - Mirian Claudia de Souza Pereira
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz. Av. Brasil 4365, Rio de Janeiro 21040-900, RJ, Brazil; (T.P.d.S.); (L.M.R.O.); (L.d.S.L.); (V.B.P.)
| |
Collapse
|
2
|
Linciano P, Quotadamo A, Luciani R, Santucci M, Zorn KM, Foil DH, Lane TR, Cordeiro da Silva A, Santarem N, B Moraes C, Freitas-Junior L, Wittig U, Mueller W, Tonelli M, Ferrari S, Venturelli A, Gul S, Kuzikov M, Ellinger B, Reinshagen J, Ekins S, Costi MP. High-Throughput Phenotypic Screening and Machine Learning Methods Enabled the Selection of Broad-Spectrum Low-Toxicity Antitrypanosomatidic Agents. J Med Chem 2023; 66:15230-15255. [PMID: 37921561 PMCID: PMC10683024 DOI: 10.1021/acs.jmedchem.3c01322] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 11/04/2023]
Abstract
Broad-spectrum anti-infective chemotherapy agents with activity against Trypanosomes, Leishmania, and Mycobacterium tuberculosis species were identified from a high-throughput phenotypic screening program of the 456 compounds belonging to the Ty-Box, an in-house industry database. Compound characterization using machine learning approaches enabled the identification and synthesis of 44 compounds with broad-spectrum antiparasitic activity and minimal toxicity against Trypanosoma brucei, Leishmania Infantum, and Trypanosoma cruzi. In vitro studies confirmed the predictive models identified in compound 40 which emerged as a new lead, featured by an innovative N-(5-pyrimidinyl)benzenesulfonamide scaffold and promising low micromolar activity against two parasites and low toxicity. Given the volume and complexity of data generated by the diverse high-throughput screening assays performed on the compounds of the Ty-Box library, the chemoinformatic and machine learning tools enabled the selection of compounds eligible for further evaluation of their biological and toxicological activities and aided in the decision-making process toward the design and optimization of the identified lead.
Collapse
Affiliation(s)
- Pasquale Linciano
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Antonio Quotadamo
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Rosaria Luciani
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Matteo Santucci
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Kimberley M. Zorn
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Daniel H. Foil
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Thomas R. Lane
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Anabela Cordeiro da Silva
- Institute
for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto
de Investigaçao e Inovaçao em Saúde, Universidade do Porto and Institute for Molecular
and Cell Biology, 4150-180 Porto, Portugal
| | - Nuno Santarem
- Institute
for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto
de Investigaçao e Inovaçao em Saúde, Universidade do Porto and Institute for Molecular
and Cell Biology, 4150-180 Porto, Portugal
| | - Carolina B Moraes
- Brazilian
Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, São Paulo, Brazil
| | - Lucio Freitas-Junior
- Brazilian
Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, São Paulo, Brazil
| | - Ulrike Wittig
- Scientific
Databases and Visualization Group and Molecular and Cellular Modelling
Group, Heidelberg Institute for Theoretical
Studies (HITS), D-69118 Heidelberg, Germany
| | - Wolfgang Mueller
- Scientific
Databases and Visualization Group and Molecular and Cellular Modelling
Group, Heidelberg Institute for Theoretical
Studies (HITS), D-69118 Heidelberg, Germany
| | - Michele Tonelli
- Department
of Pharmacy, University of Genoa, Viale Benedetto XV n.3, 16132 Genoa, Italy
| | - Stefania Ferrari
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Alberto Venturelli
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
- TYDOCK
PHARMA S.r.l., Strada
Gherbella 294/b, 41126 Modena, Italy
| | - Sheraz Gul
- Fraunhofer
Translational Medicine and Pharmacology, Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases
CIMD, Schnackenburgallee
114, D-22525 Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer
Translational Medicine and Pharmacology, Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases
CIMD, Schnackenburgallee
114, D-22525 Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer
Translational Medicine and Pharmacology, Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases
CIMD, Schnackenburgallee
114, D-22525 Hamburg, Germany
| | - Jeanette Reinshagen
- Fraunhofer
Translational Medicine and Pharmacology, Schnackenburgallee 114, D-22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases
CIMD, Schnackenburgallee
114, D-22525 Hamburg, Germany
| | - Sean Ekins
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Maria Paola Costi
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| |
Collapse
|
3
|
Panecka-Hofman J, Poehner I, Wade R. Anti-trypanosomatid structure-based drug design - lessons learned from targeting the folate pathway. Expert Opin Drug Discov 2022; 17:1029-1045. [PMID: 36073204 DOI: 10.1080/17460441.2022.2113776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Trypanosomatidic parasitic infections of humans and animals caused by Trypanosoma brucei, Trypanosoma cruzi, and Leishmania species pose a significant health and economic burden in developing countries. There are few effective and accessible treatments for these diseases, and the existing therapies suffer from problems such as parasite resistance and side effects. Structure-based drug design (SBDD) is one of the strategies that has been applied to discover new compounds targeting trypanosomatid-borne diseases. AREAS COVERED We review the current literature (mostly over the last 5 years, searched in PubMed database on Nov 11th 2021) on the application of structure-based drug design approaches to identify new anti-trypanosomatidic compounds that interfere with a validated target biochemical pathway, the trypanosomatid folate pathway. EXPERT OPINION The application of structure-based drug design approaches to perturb the trypanosomatid folate pathway has successfully provided many new inhibitors with good selectivity profiles, most of which are natural products or their derivatives or have scaffolds of known drugs. However, the inhibitory effect against the target protein(s) often does not translate to anti-parasitic activity. Further progress is hampered by our incomplete understanding of parasite biology and biochemistry, which is necessary to complement SBDD in a multiparameter optimization approach to discovering selective anti-parasitic drugs.
Collapse
Affiliation(s)
- Joanna Panecka-Hofman
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Pasteura 5a, 02-097 Warsaw, Poland
| | - Ina Poehner
- School of Pharmacy, University of Eastern Finland, Kuopio, Yliopistonranta 1C, PO Box 1627, FI-70211 Kuopio, Finland
| | - Rebecca Wade
- Center for Molecular Biology (ZMBH), Heidelberg University, Im Neuenheimer Feld 282, Heidelberg 69120, Germany.,Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, Heidelberg 69118, Germany.,DKFZ-ZMBH Alliance and Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Im Neuenheimer Feld 205, Heidelberg 69120, Germany
| |
Collapse
|
4
|
Pöhner I, Quotadamo A, Panecka-Hofman J, Luciani R, Santucci M, Linciano P, Landi G, Di Pisa F, Dello Iacono L, Pozzi C, Mangani S, Gul S, Witt G, Ellinger B, Kuzikov M, Santarem N, Cordeiro-da-Silva A, Costi MP, Venturelli A, Wade RC. Multitarget, Selective Compound Design Yields Potent Inhibitors of a Kinetoplastid Pteridine Reductase 1. J Med Chem 2022; 65:9011-9033. [PMID: 35675511 PMCID: PMC9289884 DOI: 10.1021/acs.jmedchem.2c00232] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
The optimization
of compounds with multiple targets is a difficult
multidimensional problem in the drug discovery cycle. Here, we present
a systematic, multidisciplinary approach to the development of selective
antiparasitic compounds. Computational fragment-based design of novel
pteridine derivatives along with iterations of crystallographic structure
determination allowed for the derivation of a structure–activity
relationship for multitarget inhibition. The approach yielded compounds
showing apparent picomolar inhibition of T. brucei pteridine reductase 1 (PTR1), nanomolar inhibition of L.
major PTR1, and selective submicromolar inhibition of parasite
dihydrofolate reductase (DHFR) versus human DHFR. Moreover, by combining
design for polypharmacology with a property-based on-parasite optimization,
we found three compounds that exhibited micromolar EC50 values against T. brucei brucei while retaining
their target inhibition. Our results provide a basis for the further
development of pteridine-based compounds, and we expect our multitarget
approach to be generally applicable to the design and optimization
of anti-infective agents.
Collapse
Affiliation(s)
- Ina Pöhner
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), D-69118 Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, D-69120 Heidelberg, Germany
| | - Antonio Quotadamo
- Tydock Pharma srl, Strada Gherbella 294/B, 41126 Modena, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Joanna Panecka-Hofman
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), D-69118 Heidelberg, Germany.,Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
| | - Rosaria Luciani
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Matteo Santucci
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Pasquale Linciano
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Giacomo Landi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Flavio Di Pisa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Lucia Dello Iacono
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Stefano Mangani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Gesa Witt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Nuno Santarem
- Instituto de Investigação e Inovação em Saúde, Institute for Molecular and Cell Biology, Universidade do Porto, 4200-135 Porto, Portugal
| | - Anabela Cordeiro-da-Silva
- Instituto de Investigação e Inovação em Saúde, Institute for Molecular and Cell Biology, Universidade do Porto, 4200-135 Porto, Portugal.,Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Maria P Costi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Alberto Venturelli
- Tydock Pharma srl, Strada Gherbella 294/B, 41126 Modena, Italy.,Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Rebecca C Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), D-69118 Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, D-69120 Heidelberg, Germany.,Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, D-69120 Heidelberg, Germany.,Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, D-69120 Heidelberg, Germany
| |
Collapse
|
5
|
Dello Iacono L, Di Pisa F, Mangani S. Crystal structure of the ternary complex of Leishmania major pteridine reductase 1 with the cofactor NADP +/NADPH and the substrate folic acid. Acta Crystallogr F Struct Biol Commun 2022; 78:170-176. [PMID: 35400669 PMCID: PMC8996148 DOI: 10.1107/s2053230x22002795] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/14/2022] [Indexed: 11/10/2022] Open
Abstract
Pteridine reductase 1 (PTR1) is a key enzyme of the folate pathway in protozoan parasites of the genera Leishmania and Trypanosoma and is a valuable drug target for tropical diseases. This enzyme is able to catalyze the NADPH-dependent reduction of both conjugated (folate) and unconjugated (biopterin) pterins to their tetrahydro forms, starting from oxidized- or dihydro-state substrates. The currently available X-ray structures of Leishmania major PTR1 (LmPTR1) show the enzyme in its unbound, unconjugated substrate-bound (with biopterin derivatives) and inhibitor-bound forms. However, no structure has yet been determined of LmPTR1 bound to a conjugated substrate. Here, the high-resolution crystal structure of LmPTR1 in complex with folic acid is presented and the intermolecular forces that drive the binding of the substrate in the catalytic pocket are described. By expanding the collection of LmPTR1 structures in complex with process intermediates, additional insights into the active-site rearrangements that occur during the catalytic process are provided. In contrast to previous structures with biopterin derivatives, a small but significant difference in the orientation of Asp181 and Tyr194 of the catalytic triad is found. This feature is shared by PTR1 from T. brucei (TbPTR1) in complex with the same substrate molecule and may be informative in deciphering the importance of such residues at the beginning of the catalytic process.
Collapse
Affiliation(s)
- Lucia Dello Iacono
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Flavio Di Pisa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Stefano Mangani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
6
|
Santucci M, Luciani R, Gianquinto E, Pozzi C, Pisa FD, dello Iacono L, Landi G, Tagliazucchi L, Mangani S, Spyrakis F, Costi MP. Repurposing the Trypanosomatidic GSK Kinetobox for the Inhibition of Parasitic Pteridine and Dihydrofolate Reductases. Pharmaceuticals (Basel) 2021; 14:ph14121246. [PMID: 34959646 PMCID: PMC8704748 DOI: 10.3390/ph14121246] [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: 10/31/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 11/20/2022] Open
Abstract
Three open-source anti-kinetoplastid chemical boxes derived from a whole-cell phenotypic screening by GlaxoSmithKline (Tres Cantos Anti-Kinetoplastid Screening, TCAKS) were exploited for the discovery of a novel core structure inspiring new treatments of parasitic diseases targeting the trypansosmatidic pteridine reductase 1 (PTR1) and dihydrofolate reductase (DHFR) enzymes. In total, 592 compounds were tested through medium-throughput screening assays. A subset of 14 compounds successfully inhibited the enzyme activity in the low micromolar range of at least one of the enzymes from both Trypanosoma brucei and Lesihmania major parasites (pan-inhibitors), or from both PTR1 and DHFR-TS of the same parasite (dual inhibitors). Molecular docking studies of the protein–ligand interaction focused on new scaffolds not reproducing the well-known antifolate core clearly explaining the experimental data. TCMDC-143249, classified as a benzenesulfonamide derivative by the QikProp descriptor tool, showed selective inhibition of PTR1 and growth inhibition of the kinetoplastid parasites in the 5 μM range. In our work, we enlarged the biological profile of the GSK Kinetobox and identified new core structures inhibiting selectively PTR1, effective against the kinetoplastid infectious protozoans. In perspective, we foresee the development of selective PTR1 and DHFR inhibitors for studies of drug combinations.
Collapse
Affiliation(s)
- Matteo Santucci
- Department of Life Science, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy; (M.S.); (R.L.); (L.T.)
| | - Rosaria Luciani
- Department of Life Science, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy; (M.S.); (R.L.); (L.T.)
| | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Turin, Italy; (E.G.); (F.S.)
| | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy—Department of Excellence 2018–2020, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.P.); (F.d.P.); (L.d.I.); (G.L.); (S.M.)
| | - Flavio di Pisa
- Department of Biotechnology, Chemistry and Pharmacy—Department of Excellence 2018–2020, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.P.); (F.d.P.); (L.d.I.); (G.L.); (S.M.)
| | - Lucia dello Iacono
- Department of Biotechnology, Chemistry and Pharmacy—Department of Excellence 2018–2020, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.P.); (F.d.P.); (L.d.I.); (G.L.); (S.M.)
| | - Giacomo Landi
- Department of Biotechnology, Chemistry and Pharmacy—Department of Excellence 2018–2020, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.P.); (F.d.P.); (L.d.I.); (G.L.); (S.M.)
| | - Lorenzo Tagliazucchi
- Department of Life Science, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy; (M.S.); (R.L.); (L.T.)
| | - Stefano Mangani
- Department of Biotechnology, Chemistry and Pharmacy—Department of Excellence 2018–2020, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.P.); (F.d.P.); (L.d.I.); (G.L.); (S.M.)
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, Via Giuria 9, 10125 Turin, Italy; (E.G.); (F.S.)
| | - Maria Paola Costi
- Department of Life Science, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy; (M.S.); (R.L.); (L.T.)
- Correspondence:
| |
Collapse
|
7
|
Kokovina TS, Gadomsky SY, Terentiev AA, Sanina NA. A Novel Approach to the Synthesis of 1,3,4-Thiadiazole-2-amine Derivatives. Molecules 2021; 26:molecules26175159. [PMID: 34500593 PMCID: PMC8434302 DOI: 10.3390/molecules26175159] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
The main purpose of the study was the development of a new method for synthesis of 1,3,4-thiadiazol-2-amine derivatives in a one-pot manner using the reaction between a thiosemicarbazide and carboxylic acid without toxic additives such as POCl3 or SOCl2. The reaction was investigated in the presence of polyphosphate ester (PPE). It was found that, in the presence of PPE, the reaction between the thiosemicarbazide and carboxylic acid proceeds in one-pot through three steps with the formation of corresponding 2-amino-1,3,4-thiadiazole. Using the developed approach five, 2-amino-1,3,4-thiadiazoles were synthesized. The structures of all compounds were proven by mass spectrometry, IR, and NMR spectroscopies.
Collapse
Affiliation(s)
- Tatiana S. Kokovina
- Institute of Problems of Chemical Physics, The Russian Academy of Sciences, 142432 Chernogolovka, Russia; (T.S.K.); (A.A.T.); (N.A.S.)
- Faculty of Fundamental Physical and Chemical Engineering, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Svyatoslav Y. Gadomsky
- Institute of Problems of Chemical Physics, The Russian Academy of Sciences, 142432 Chernogolovka, Russia; (T.S.K.); (A.A.T.); (N.A.S.)
- Correspondence:
| | - Alexei A. Terentiev
- Institute of Problems of Chemical Physics, The Russian Academy of Sciences, 142432 Chernogolovka, Russia; (T.S.K.); (A.A.T.); (N.A.S.)
- Faculty of Fundamental Physical and Chemical Engineering, Lomonosov Moscow State University, 119991 Moscow, Russia
- Medicinal Chemistry Science and Education Center, Moscow Region State University, 141014 Mytishchi, Russia
| | - Nataliya A. Sanina
- Institute of Problems of Chemical Physics, The Russian Academy of Sciences, 142432 Chernogolovka, Russia; (T.S.K.); (A.A.T.); (N.A.S.)
- Faculty of Fundamental Physical and Chemical Engineering, Lomonosov Moscow State University, 119991 Moscow, Russia
- Medicinal Chemistry Science and Education Center, Moscow Region State University, 141014 Mytishchi, Russia
| |
Collapse
|
8
|
Tassone G, Landi G, Linciano P, Francesconi V, Tonelli M, Tagliazucchi L, Costi MP, Mangani S, Pozzi C. Evidence of Pyrimethamine and Cycloguanil Analogues as Dual Inhibitors of Trypanosoma brucei Pteridine Reductase and Dihydrofolate Reductase. Pharmaceuticals (Basel) 2021; 14:636. [PMID: 34209148 PMCID: PMC8308740 DOI: 10.3390/ph14070636] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
Trypanosoma and Leishmania parasites are the etiological agents of various threatening neglected tropical diseases (NTDs), including human African trypanosomiasis (HAT), Chagas disease, and various types of leishmaniasis. Recently, meaningful progresses in the treatment of HAT, due to Trypanosoma brucei (Tb), have been achieved by the introduction of fexinidazole and the combination therapy eflornithine-nifurtimox. Nevertheless, due to drug resistance issues and the exitance of animal reservoirs, the development of new NTD treatments is still required. For this purpose, we explored the combined targeting of two key folate enzymes, dihydrofolate reductase (DHFR) and pteridine reductase 1 (PTR1). We formerly showed that the TbDHFR inhibitor cycloguanil (CYC) also targets TbPTR1, although with reduced affinity. Here, we explored a small library of CYC analogues to understand how their substitution pattern affects the inhibition of both TbPTR1 and TbDHFR. Some novel structural features responsible for an improved, but preferential, ability of CYC analogues to target TbPTR1 were disclosed. Furthermore, we showed that the known drug pyrimethamine (PYR) effectively targets both enzymes, also unveiling its binding mode to TbPTR1. The structural comparison between PYR and CYC binding modes to TbPTR1 and TbDHFR provided key insights for the future design of dual inhibitors for HAT therapy.
Collapse
Affiliation(s)
- Giusy Tassone
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, via Aldo Moro 2, 53100 Siena, Italy; (G.T.); (G.L.); (S.M.)
| | - Giacomo Landi
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, via Aldo Moro 2, 53100 Siena, Italy; (G.T.); (G.L.); (S.M.)
| | - Pasquale Linciano
- Department of Life Science, University of Modena and Reggio Emilia, via Campi 103, 41125 Modena, Italy; (P.L.); (L.T.); (M.P.C.)
| | - Valeria Francesconi
- Department of Pharmacy, University of Genoa, Viale Benedetto XV n.3, 16132 Genoa, Italy; (V.F.); (M.T.)
| | - Michele Tonelli
- Department of Pharmacy, University of Genoa, Viale Benedetto XV n.3, 16132 Genoa, Italy; (V.F.); (M.T.)
| | - Lorenzo Tagliazucchi
- Department of Life Science, University of Modena and Reggio Emilia, via Campi 103, 41125 Modena, Italy; (P.L.); (L.T.); (M.P.C.)
| | - Maria Paola Costi
- Department of Life Science, University of Modena and Reggio Emilia, via Campi 103, 41125 Modena, Italy; (P.L.); (L.T.); (M.P.C.)
| | - Stefano Mangani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, via Aldo Moro 2, 53100 Siena, Italy; (G.T.); (G.L.); (S.M.)
| | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018–2022, University of Siena, via Aldo Moro 2, 53100 Siena, Italy; (G.T.); (G.L.); (S.M.)
| |
Collapse
|
9
|
Toward Chemical Validation of Leishmania infantum Ribose 5-Phosphate Isomerase as a Drug Target. Antimicrob Agents Chemother 2021; 65:e0189220. [PMID: 33875438 DOI: 10.1128/aac.01892-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neglected tropical diseases caused by kinetoplastid parasites (Trypanosoma brucei, Trypanosoma cruzi, and Leishmania spp.) place a significant health and economic burden on developing nations worldwide. Current therapies are largely outdated, inadequate, and face mounting drug resistance from the causative parasites. Thus, there is an urgent need for drug discovery and development. Target-led drug discovery approaches have focused on the identification of parasite enzymes catalyzing essential biochemical processes, which significantly differ from equivalent proteins found in humans, thereby providing potentially exploitable therapeutic windows. One such target is ribose 5-phosphate isomerase B (RpiB), an enzyme involved in the nonoxidative branch of the pentose phosphate pathway, which catalyzes the interconversion of d-ribose 5-phosphate and d-ribulose 5-phosphate. Although protozoan RpiB has been the focus of numerous targeted studies, compounds capable of selectively inhibiting this parasite enzyme have not been identified. Here, we present the results of a fragment library screening against Leishmania infantum RpiB (LiRpiB), performed using thermal shift analysis. Hit fragments were shown to be effective inhibitors of LiRpiB in activity assays, and several fragments were capable of selectively inhibiting parasite growth in vitro. These results support the identification of LiRpiB as a validated therapeutic target. The X-ray crystal structure of apo LiRpiB was also solved, permitting docking studies to assess how hit fragments might interact with LiRpiB to inhibit its activity. Overall, this work will guide structure-based development of LiRpiB inhibitors as antileishmanial agents.
Collapse
|
10
|
2-Amino-1,3,4-thiadiazoles as prospective agents in trypanosomiasis and other parasitoses. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2020; 70:259-290. [PMID: 32074064 DOI: 10.2478/acph-2020-0031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 01/19/2023]
Abstract
Parasitic diseases are a serious public health problem affecting hundreds of millions of people worldwide. African trypanosomiasis, American trypanosomiasis, leishmaniasis, malaria and toxoplasmosis are the main parasitic infections caused by protozoan parasites with over one million deaths each year. Due to old medications and drug resistance worldwide, there is an urgent need for new antiparasitic drugs. 1,3,4-Thiadiazoles have been widely studied for medical applications. The chemical, physical and pharmacokinetic properties recommend 1,3,4-thiadiazole ring as a target in drug development. Many scientific papers report the antiparasitic potential of 2-amino-1,3,4-thiadiazoles. This review presents synthetic 2-amino-1,3,4-thiadiazoles exhibiting antitrypanosomal, antimalarial and antitoxoplasmal activities. Although there are insufficient results to state the quality of 2-amino-1,3,4-thiadiazoles as a new class of antiparasitic agents, many reported derivatives can be considered as lead compounds for drug synthesis and a promise for the future treatment of parasitosis and provide a valid strategy for the development of potent antiparasitic drugs.
Collapse
|
11
|
Landi G, Linciano P, Tassone G, Costi MP, Mangani S, Pozzi C. High-resolution crystal structure of Trypanosoma brucei pteridine reductase 1 in complex with an innovative tricyclic-based inhibitor. Acta Crystallogr D Struct Biol 2020; 76:558-564. [PMID: 32496217 DOI: 10.1107/s2059798320004891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/06/2020] [Indexed: 11/10/2022] Open
Abstract
The protozoan parasite Trypanosoma brucei is the etiological agent of human African trypanosomiasis (HAT). HAT, together with other neglected tropical diseases, causes serious health and economic issues, especially in tropical and subtropical areas. The classical antifolates targeting dihydrofolate reductase (DHFR) are ineffective towards trypanosomatid parasites owing to a metabolic bypass by the expression of pteridine reductase 1 (PTR1). The combined inhibition of PTR1 and DHFR activities in Trypanosoma parasites represents a promising strategy for the development of new effective treatments for HAT. To date, only monocyclic and bicyclic aromatic systems have been proposed as inhibitors of T. brucei PTR1 (TbPTR1); nevertheless, the size of the catalytic cavity allows the accommodation of expanded molecular cores. Here, an innovative tricyclic-based compound has been explored as a TbPTR1-targeting molecule and its potential application for the development of a new class of PTR1 inhibitors has been evaluated. 2,4-Diaminopyrimido[4,5-b]indol-6-ol (1) was designed and synthesized, and was found to be effective in blocking TbPTR1 activity, with a Ki in the low-micromolar range. The binding mode of 1 was clarified through the structural characterization of its ternary complex with TbPTR1 and the cofactor NADP(H), which was determined to 1.30 Å resolution. The compound adopts a substrate-like orientation inside the cavity that maximizes the binding contributions of hydrophobic and hydrogen-bond interactions. The binding mode of 1 was compared with those of previously reported bicyclic inhibitors, providing new insights for the design of innovative tricyclic-based molecules targeting TbPTR1.
Collapse
Affiliation(s)
- Giacomo Landi
- Department of Biotechnology, Chemistry and Pharmacy - Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Pasquale Linciano
- Department of Life Science, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Giusy Tassone
- Department of Biotechnology, Chemistry and Pharmacy - Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Maria Paola Costi
- Department of Life Science, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Stefano Mangani
- Department of Biotechnology, Chemistry and Pharmacy - Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy - Department of Excellence 2018-2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
12
|
Shaykoon MS, Marzouk AA, Soltan OM, Wanas AS, Radwan MM, Gouda AM, Youssif BGM, Abdel-Aziz M. Design, synthesis and antitrypanosomal activity of heteroaryl-based 1,2,4-triazole and 1,3,4-oxadiazole derivatives. Bioorg Chem 2020; 100:103933. [PMID: 32446119 DOI: 10.1016/j.bioorg.2020.103933] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/10/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
Two series of novel 1,2,4-triazol-3-yl-thioacetamide 3a-b and 4a-b and 5-pyrazin-2-yl-3H-[1,3,4]oxadiazole-2-thiones 9a-h were designed and synthesized. The compounds prepared have been identified using 1H NMR, 13C NMR and elemental analyses. The synthesized compounds 3a, 3b, 4a, 4b, 9a, 9b, 9d-e and 9f have been evaluated with α-difluoromethylornithine (DFMO) as a control drug for their in vitro antitrypanosomal activity against Trypanosoma brucei. Results showed that 3b was the most active compound in general and also more potent than control DFMO. 3b was 8 folds more potent than the reference with IC50 of 0.79 μM and IC90 of 1.35 μM, respectively compared to DFMO (IC50 = 6.10 μM and IC90 of 8.66 μM). The tested compounds showed moderate cytotoxicity with selectivity indices ranging from 12 (9d) to 102 (3b) against L6 cells. Docking study was performed into ten of T. brucei enzymes which have been identified as potential/valid targets for most of the antitrypanosomal agents. The results of the docking study revealed high binding scores toward many of the selected enzymes. A good correlation was observed only between log (IC50) of antitrypanosomal activity of the new compounds and their calculated Ki values against TryR enzyme (R2 = 0.726). Compound 3b, the most active as antitrypanosomal agents exhibited similar binding orientation and interaction to those of WP6 against TryR enzyme. However, in a next round of work, a complementary studies will be carried out to clarify the mechanism of action of these compounds.
Collapse
Affiliation(s)
- Montaser Sh Shaykoon
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Adel A Marzouk
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Osama M Soltan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Amira S Wanas
- National Center for Natural Products Research, University of Mississippi, MS 38677, USA; Pharmacognosy Department, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt
| | - Mohamed M Radwan
- National Center for Natural Products Research, University of Mississippi, MS 38677, USA
| | - Ahmed M Gouda
- Department of Medicinal Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Bahaa G M Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| | - Mohamed Abdel-Aziz
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519 Minia, Egypt.
| |
Collapse
|
13
|
Moraes CB, Witt G, Kuzikov M, Ellinger B, Calogeropoulou T, Prousis KC, Mangani S, Di Pisa F, Landi G, Iacono LD, Pozzi C, Freitas-Junior LH, Dos Santos Pascoalino B, Bertolacini CP, Behrens B, Keminer O, Leu J, Wolf M, Reinshagen J, Cordeiro-da-Silva A, Santarem N, Venturelli A, Wrigley S, Karunakaran D, Kebede B, Pöhner I, Müller W, Panecka-Hofman J, Wade RC, Fenske M, Clos J, Alunda JM, Corral MJ, Uliassi E, Bolognesi ML, Linciano P, Quotadamo A, Ferrari S, Santucci M, Borsari C, Costi MP, Gul S. Accelerating Drug Discovery Efforts for Trypanosomatidic Infections Using an Integrated Transnational Academic Drug Discovery Platform. SLAS DISCOVERY 2020; 24:346-361. [PMID: 30784368 PMCID: PMC6484532 DOI: 10.1177/2472555218823171] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
According to the World Health Organization, more than 1 billion people are at risk of or are affected by neglected tropical diseases. Examples of such diseases include trypanosomiasis, which causes sleeping sickness; leishmaniasis; and Chagas disease, all of which are prevalent in Africa, South America, and India. Our aim within the New Medicines for Trypanosomatidic Infections project was to use (1) synthetic and natural product libraries, (2) screening, and (3) a preclinical absorption, distribution, metabolism, and excretion-toxicity (ADME-Tox) profiling platform to identify compounds that can enter the trypanosomatidic drug discovery value chain. The synthetic compound libraries originated from multiple scaffolds with known antiparasitic activity and natural products from the Hypha Discovery MycoDiverse natural products library. Our focus was first to employ target-based screening to identify inhibitors of the protozoan Trypanosoma brucei pteridine reductase 1 ( TbPTR1) and second to use a Trypanosoma brucei phenotypic assay that made use of the T. brucei brucei parasite to identify compounds that inhibited cell growth and caused death. Some of the compounds underwent structure-activity relationship expansion and, when appropriate, were evaluated in a preclinical ADME-Tox assay panel. This preclinical platform has led to the identification of lead-like compounds as well as validated hits in the trypanosomatidic drug discovery value chain.
Collapse
Affiliation(s)
- Carolina B Moraes
- 1 Laboratório Nacional de Biociências (LNBio), Centro de Pesquisa em Energia e Materiais (CNPEM), Campinas-SP, Brazil.,2 Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo-SP, Brazil
| | - Gesa Witt
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Maria Kuzikov
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Bernhard Ellinger
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Theodora Calogeropoulou
- 4 National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, Athens, Greece
| | - Kyriakos C Prousis
- 4 National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, Athens, Greece
| | - Stefano Mangani
- 5 Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Flavio Di Pisa
- 5 Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Giacomo Landi
- 5 Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Lucia Dello Iacono
- 5 Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Cecilia Pozzi
- 5 Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Lucio H Freitas-Junior
- 1 Laboratório Nacional de Biociências (LNBio), Centro de Pesquisa em Energia e Materiais (CNPEM), Campinas-SP, Brazil.,2 Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo-SP, Brazil
| | - Bruno Dos Santos Pascoalino
- 1 Laboratório Nacional de Biociências (LNBio), Centro de Pesquisa em Energia e Materiais (CNPEM), Campinas-SP, Brazil
| | - Claudia P Bertolacini
- 1 Laboratório Nacional de Biociências (LNBio), Centro de Pesquisa em Energia e Materiais (CNPEM), Campinas-SP, Brazil
| | - Birte Behrens
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Oliver Keminer
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Jennifer Leu
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Markus Wolf
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Jeanette Reinshagen
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| | - Anabela Cordeiro-da-Silva
- 6 Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, Porto, Portugal
| | - Nuno Santarem
- 6 Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, Porto, Portugal
| | | | | | | | | | - Ina Pöhner
- 9 Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Wolfgang Müller
- 9 Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Joanna Panecka-Hofman
- 9 Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany.,11 Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Rebecca C Wade
- 9 Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany.,12 Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany.,13 Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Heidelberg, Germany
| | - Martina Fenske
- 14 Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - Joachim Clos
- 15 Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | - Elisa Uliassi
- 17 Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | - Pasquale Linciano
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio Quotadamo
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Stefania Ferrari
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Santucci
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Borsari
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Maria Paola Costi
- 18 Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sheraz Gul
- 3 Fraunhofer Institute for Molecular Biology and Applied Ecology-ScreeningPort, Hamburg, Germany
| |
Collapse
|
14
|
Further insight into the dual COX-2 and 15-LOX anti-inflammatory activity of 1,3,4-thiadiazole-thiazolidinone hybrids: The contribution of the substituents at 5th positions is size dependent. Bioorg Chem 2020; 97:103657. [DOI: 10.1016/j.bioorg.2020.103657] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/28/2020] [Accepted: 02/09/2020] [Indexed: 12/16/2022]
|
15
|
Linciano P, Cullia G, Borsari C, Santucci M, Ferrari S, Witt G, Gul S, Kuzikov M, Ellinger B, Santarém N, Cordeiro da Silva A, Conti P, Bolognesi ML, Roberti M, Prati F, Bartoccini F, Retini M, Piersanti G, Cavalli A, Goldoni L, Bertozzi SM, Bertozzi F, Brambilla E, Rizzo V, Piomelli D, Pinto A, Bandiera T, Costi MP. Identification of a 2,4-diaminopyrimidine scaffold targeting Trypanosoma brucei pteridine reductase 1 from the LIBRA compound library screening campaign. Eur J Med Chem 2020; 189:112047. [PMID: 31982652 DOI: 10.1016/j.ejmech.2020.112047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/31/2019] [Accepted: 01/06/2020] [Indexed: 12/21/2022]
Abstract
The LIBRA compound library is a collection of 522 non-commercial molecules contributed by various Italian academic laboratories. These compounds have been designed and synthesized during different medicinal chemistry programs and are hosted by the Italian Institute of Technology. We report the screening of the LIBRA compound library against Trypanosoma brucei and Leishmania major pteridine reductase 1, TbPTR1 and LmPTR1. Nine compounds were active against parasitic PTR1 and were selected for cell-based parasite screening, as single agents and in combination with methotrexate (MTX). The most interesting TbPTR1 inhibitor identified was 4-(benzyloxy)pyrimidine-2,6-diamine (LIB_66). Subsequently, six new LIB_66 derivatives were synthesized to explore its Structure-Activity-Relationship (SAR) and absorption, distribution, metabolism, excretion and toxicity (ADMET) properties. The results indicate that PTR1 has a preference to bind inhibitors, which resemble its biopterin/folic acid substrates, such as the 2,4-diaminopyrimidine derivatives.
Collapse
Affiliation(s)
- Pasquale Linciano
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Gregorio Cullia
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133, Milan, Italy
| | - Chiara Borsari
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Matteo Santucci
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Stefania Ferrari
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology and Applied Ecology - ScreeningPort, Hamburg, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology - ScreeningPort, Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Molecular Biology and Applied Ecology - ScreeningPort, Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology - ScreeningPort, Hamburg, Germany
| | - Nuno Santarém
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal and Instituto de Investigação e Inovação Em Saúde, Universidade Do Porto, 4150-180, Porto, Portugal
| | - Anabela Cordeiro da Silva
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal and Instituto de Investigação e Inovação Em Saúde, Universidade Do Porto, 4150-180, Porto, Portugal; Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Portugal
| | - Paola Conti
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133, Milan, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126, Bologna, Italy
| | - Marinella Roberti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126, Bologna, Italy
| | - Federica Prati
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126, Bologna, Italy
| | - Francesca Bartoccini
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Michele Retini
- Department of Biomolecular Sciences, Section of Chemistry, University of Urbino "Carlo Bo", Piazza Rinascimento 6, 61029, Urbino, Italy
| | - Giovanni Piersanti
- Department of Biomolecular Sciences, Section of Chemistry, University of Urbino "Carlo Bo", Piazza Rinascimento 6, 61029, Urbino, Italy
| | - Andrea Cavalli
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126, Bologna, Italy; Computational and Chemical Biology, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Luca Goldoni
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Sine Mandrup Bertozzi
- Analytical Chemistry Lab, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Fabio Bertozzi
- PharmaChemistry Line, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Enzo Brambilla
- PharmaChemistry Line, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Vincenzo Rizzo
- PharmaChemistry Line, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Daniele Piomelli
- Departments of Anatomy and Neurobiology, Pharmacology and Biological Chemistry, University of California, Irvine, 92697-4625, USA
| | - Andrea Pinto
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133, Milan, Italy
| | - Tiziano Bandiera
- PharmaChemistry Line, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Maria Paola Costi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, 41125, Modena, Italy.
| |
Collapse
|
16
|
Teixeira BVF, Teles ALB, da Silva SG, Brito CCB, de Freitas HF, Pires ABL, Froes TQ, Castilho MS. Dual and selective inhibitors of pteridine reductase 1 (PTR1) and dihydrofolate reductase-thymidylate synthase (DHFR-TS) from Leishmania chagasi. J Enzyme Inhib Med Chem 2019; 34:1439-1450. [PMID: 31409157 PMCID: PMC6713189 DOI: 10.1080/14756366.2019.1651311] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/27/2019] [Accepted: 07/29/2019] [Indexed: 02/06/2023] Open
Abstract
Leishmaniasis is a tropical disease found in more than 90 countries. The drugs available to treat this disease have nonspecific action and high toxicity. In order to develop novel therapeutic alternatives to fight this ailment, pteridine reductase 1 (PTR1) and dihydrofolate reductase-thymidylate synthase (DHF-TS) have been targeted, once Leishmania is auxotrophic for folates. Although PTR1 and DHFR-TS from other protozoan parasites have been studied, their homologs in Leishmania chagasi have been poorly characterized. Hence, this work describes the optimal conditions to express the recombinant LcPTR1 and LcDHFR-TS enzymes, as well as balanced assay conditions for screening. Last but not the least, we show that 2,4 diaminopyrimidine derivatives are low-micromolar competitive inhibitors of both enzymes (LcPTR1 Ki = 1.50-2.30 µM and LcDHFR Ki = 0.28-3.00 µM) with poor selectivity index. On the other hand, compound 5 (2,4-diaminoquinazoline derivative) is a selective LcPTR1 inhibitor (Ki = 0.47 µM, selectivity index = 20).
Collapse
Affiliation(s)
| | - André Lacerda Braga Teles
- Programa de Pós-Graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Estadual da Bahia, Salvador, BA, Brazil
- Departamento de Ciências da Vida, Universidade do Estado da Bahia, Salvador, BA, Brazil
| | | | | | - Humberto Fonseca de Freitas
- Programa de Pós-Graduação em Farmácia, Universidade Federal da Bahia, Salvador, BA, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
| | | | - Thamires Quadros Froes
- Programa de Pós-Graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
| | - Marcelo Santos Castilho
- Programa de Pós-Graduação em Farmácia, Universidade Federal da Bahia, Salvador, BA, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Estadual de Feira de Santana, Feira de Santana, BA, Brazil
| |
Collapse
|
17
|
Activation of Caspase-6 Is Promoted by a Mutant Huntingtin Fragment and Blocked by an Allosteric Inhibitor Compound. Cell Chem Biol 2019; 26:1295-1305.e6. [PMID: 31353319 DOI: 10.1016/j.chembiol.2019.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 01/08/2019] [Accepted: 06/28/2019] [Indexed: 01/04/2023]
Abstract
Aberrant activation of caspase-6 (C6) in the absence of other hallmarks of apoptosis has been demonstrated in cells and tissues from patients with Huntington disease (HD) and animal models. C6 activity correlates with disease progression in patients with HD and the cleavage of mutant huntingtin (mHTT) protein is thought to strongly contribute to disease pathogenesis. Here we show that the mHTT1-586 fragment generated by C6 cleavage interacts with the zymogen form of the enzyme, stabilizing a conformation that contains an active site and is prone to full activation. This shift toward enhanced activity can be prevented by a small-molecule inhibitor that blocks the interaction between C6 and mHTT1-586. Molecular docking studies suggest that the inhibitor binds an allosteric site in the C6 zymogen. The interaction of mHTT1-586 with C6 may therefore promote a self-reinforcing, feedforward cycle of C6 zymogen activation and mHTT cleavage driving HD pathogenesis.
Collapse
|
18
|
Targeting pteridine reductase 1 and dihydrofolate reductase: the old is a new trend for leishmaniasis drug discovery. Future Med Chem 2019; 11:2107-2130. [DOI: 10.4155/fmc-2018-0512] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Leishmaniasis is one of the major neglected tropical diseases in the world and it is considered endemic in 88 countries. This disease is transmitted by a Leishmania spp. infected sandfly and it may lead to cutaneous or systemic manifestations. The preconized treatment has low efficacy and there are cases of resistance to some drugs. Therefore, the search for new efficient molecular targets that can lead to the preparation of new drugs must be pursued. This review aims to evaluate both Leishmania enzymes PTR1 and DHFR-TS as potential drug targets, highlight their inhibitors and to discuss critically the use of chemoinformatics to elucidate interactions and propose new molecules against these enzymes.
Collapse
|
19
|
Landi G, Linciano P, Borsari C, Bertolacini CP, Moraes CB, Cordeiro-da-Silva A, Gul S, Witt G, Kuzikov M, Costi MP, Pozzi C, Mangani S. Structural Insights into the Development of Cycloguanil Derivatives as Trypanosoma brucei Pteridine-Reductase-1 Inhibitors. ACS Infect Dis 2019; 5:1105-1114. [PMID: 31012301 DOI: 10.1021/acsinfecdis.8b00358] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cycloguanil is a known dihydrofolate-reductase (DHFR) inhibitor, but there is no evidence of its activity on pteridine reductase (PTR), the main metabolic bypass to DHFR inhibition in trypanosomatid parasites. Here, we provide experimental evidence of cycloguanil as an inhibitor of Trypanosoma brucei PTR1 (TbPTR1). A small library of cycloguanil derivatives was developed, resulting in 1 and 2a having IC50 values of 692 and 186 nM, respectively, toward TbPTR1. Structural analysis revealed that the increased potency of 1 and 2a is due to the combined contributions of hydrophobic interactions, H-bonds, and halogen bonds. Moreover, in vitro cell-growth-inhibition tests indicated that 2a is also effective on T. brucei. The simultaneous inhibition of DHFR and PTR1 activity in T. brucei is a promising new strategy for the treatment of human African trypanosomiasis. For this purpose, 1,6-dihydrotriazines represent new molecular tools to develop potent dual PTR and DHFR inhibitors.
Collapse
Affiliation(s)
- Giacomo Landi
- Department of Biotechnology, Chemistry and Pharmacy—Department of Excellence 2018−2020, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Pasquale Linciano
- Department of Life Science, University of Modena and Reggio Emilia, via Campi 103, 41125 Modena, Italy
| | - Chiara Borsari
- Department of Life Science, University of Modena and Reggio Emilia, via Campi 103, 41125 Modena, Italy
| | - Claudia P. Bertolacini
- National Laboratory of Biosciences, National Center for Research in Energy and Materials, Campinas, São Paulo 13083-970, Brazil
| | - Carolina B. Moraes
- National Laboratory of Biosciences, National Center for Research in Energy and Materials, Campinas, São Paulo 13083-970, Brazil
| | - Anabela Cordeiro-da-Silva
- Instituto de Investigação e Inovação em Saúde and IBMC-Institute for Molecular and Cell Biology, Universidade do Porto and Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), 4150-180 Porto, Portugal
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology & Applied Ecology—ScreeningPort, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology & Applied Ecology—ScreeningPort, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Molecular Biology & Applied Ecology—ScreeningPort, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Maria Paola Costi
- Department of Life Science, University of Modena and Reggio Emilia, via Campi 103, 41125 Modena, Italy
| | - Cecilia Pozzi
- Department of Biotechnology, Chemistry and Pharmacy—Department of Excellence 2018−2020, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Stefano Mangani
- Department of Biotechnology, Chemistry and Pharmacy—Department of Excellence 2018−2020, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
20
|
Serban G. Future Prospects in the Treatment of Parasitic Diseases: 2-Amino-1,3,4-Thiadiazoles in Leishmaniasis. Molecules 2019; 24:E1557. [PMID: 31010226 PMCID: PMC6514673 DOI: 10.3390/molecules24081557] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/14/2019] [Accepted: 04/17/2019] [Indexed: 02/08/2023] Open
Abstract
Neglected tropical diseases affect the lives of a billion people worldwide. Among them, the parasitic infections caused by protozoan parasites of the Trypanosomatidae family have a huge impact on human health. Leishmaniasis, caused by Leishmania spp., is an endemic parasitic disease in over 88 countries and is closely associated with poverty. Although significant advances have been made in the treatment of leishmaniasis over the last decade, currently available chemotherapy is far from satisfactory. The lack of an approved vaccine, effective medication and significant drug resistance worldwide had led to considerable interest in discovering new, inexpensive, efficient and safe antileishmanial agents. 1,3,4-Thiadiazole rings are found in biologically active natural products and medicinally important synthetic compounds. The thiadiazole ring exhibits several specific properties: it is a bioisostere of pyrimidine or benzene rings with prevalence in biologically active compounds; the sulfur atom increases lipophilicity and combined with the mesoionic character of thiadiazoles imparts good oral absorption and good cell permeability, resulting in good bioavailability. This review presents synthetic 2-amino-1,3,4-thiadiazole derivatives with antileishmanial activity. Many reported derivatives can be considered as lead compounds for the synthesis of future agents as an alternative to the treatment of leishmaniasis.
Collapse
Affiliation(s)
- Georgeta Serban
- Pharmaceutical Chemistry Department, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga, 410028 Oradea, Romania.
| |
Collapse
|
21
|
Linciano P, Pozzi C, Iacono LD, di Pisa F, Landi G, Bonucci A, Gul S, Kuzikov M, Ellinger B, Witt G, Santarem N, Baptista C, Franco C, Moraes CB, Müller W, Wittig U, Luciani R, Sesenna A, Quotadamo A, Ferrari S, Pöhner I, Cordeiro-da-Silva A, Mangani S, Costantino L, Costi MP. Enhancement of Benzothiazoles as Pteridine Reductase-1 Inhibitors for the Treatment of Trypanosomatidic Infections. J Med Chem 2019; 62:3989-4012. [DOI: 10.1021/acs.jmedchem.8b02021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Pasquale Linciano
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Cecilia Pozzi
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Lucia dello Iacono
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Flavio di Pisa
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Giacomo Landi
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Alessio Bonucci
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Nuno Santarem
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Catarina Baptista
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Caio Franco
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisaem Energia e Materiais (CNPEM), 13083-100 Campinas, São Paulo, Brazil
| | - Carolina B. Moraes
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisaem Energia e Materiais (CNPEM), 13083-100 Campinas, São Paulo, Brazil
| | | | | | - Rosaria Luciani
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Antony Sesenna
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Antonio Quotadamo
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Stefania Ferrari
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | | | - Anabela Cordeiro-da-Silva
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Stefano Mangani
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Luca Costantino
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Maria Paola Costi
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| |
Collapse
|
22
|
Linciano P, Moraes CB, Alcantara LM, Franco CH, Pascoalino B, Freitas-Junior LH, Macedo S, Santarem N, Cordeiro-da-Silva A, Gul S, Witt G, Kuzikov M, Ellinger B, Ferrari S, Luciani R, Quotadamo A, Costantino L, Costi MP. Aryl thiosemicarbazones for the treatment of trypanosomatidic infections. Eur J Med Chem 2018; 146:423-434. [PMID: 29407968 DOI: 10.1016/j.ejmech.2018.01.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 01/12/2018] [Accepted: 01/14/2018] [Indexed: 01/07/2023]
Abstract
Basing on a library of thiadiazole derivatives showing anti-trypanosomatidic activity, we have considered the thiadiazoles opened forms and reaction intermediates, thiosemicarbazones, as compounds of interest for phenotypic screening against Trypanosoma brucei (Tb), intracellular amastigote form of Leishmania infantum (Li) and Trypanosoma cruzi (Tc). Similar compounds have already shown interesting activity against the same organisms. The compounds were particularly effective against T. brucei and T. cruzi. Among the 28 synthesized compounds, the best one was (E)-2-(4-((3.4-dichlorobenzyl)oxy)benzylidene) hydrazinecarbothioamide (A14) yielding a comparable anti-parasitic activity against the three parasitic species (TbEC50 = 2.31 μM, LiEC50 = 6.14 μM, TcEC50 = 1.31 μM) and a Selectivity Index higher than 10 with respect to human macrophages, therefore showing a pan-anti-trypanosomatidic activity. (E)-2-((3'.4'-dimethoxy-[1.1'-biphenyl]-3-yl)methyle ne) hydrazinecarbothioamide (A12) and (E)-2-(4-((3.4-dichlorobenzyl)oxy)benzylidene)hydrazine carbothioamide (A14) were able to potentiate the anti-parasitic activity of methotrexate (MTX) when evaluated in combination against T. brucei, yielding a 6-fold and 4-fold respectively Dose Reduction Index for MTX. The toxicity profile against four human cell lines and a panel of in vitro early-toxicity assays (comprising hERG, Aurora B, five cytochrome P450 isoforms and mitochondrial toxicity) demonstrated the low toxicity for the thosemicarbazones class in comparison with known drugs. The results confirmed thiosemicarbazones as a suitable chemical scaffold with potential for the development of properly decorated new anti-parasitic drugs.
Collapse
Affiliation(s)
- Pasquale Linciano
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Carolina B Moraes
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas, SP, Brazil
| | - Laura M Alcantara
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas, SP, Brazil
| | - Caio H Franco
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas, SP, Brazil
| | - Bruno Pascoalino
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas, SP, Brazil
| | - Lucio H Freitas-Junior
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970, Campinas, SP, Brazil
| | - Sara Macedo
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal; Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Nuno Santarem
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal; Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Anabela Cordeiro-da-Silva
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal; Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal; Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, Hamburg, Germany
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, Hamburg, Germany
| | - Stefania Ferrari
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Rosaria Luciani
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Antonio Quotadamo
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Luca Costantino
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Maria Paola Costi
- University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy.
| |
Collapse
|