1
|
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
|
2
|
Panecka-Hofman J, Poehner I. Structure and dynamics of pteridine reductase 1: the key phenomena relevant to enzyme function and drug design. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2023; 52:521-532. [PMID: 37608196 PMCID: PMC10618315 DOI: 10.1007/s00249-023-01677-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 07/08/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023]
Abstract
Pteridine reductase 1 (PTR1) is a folate and pterin pathway enzyme unique for pathogenic trypanosomatids. As a validated drug target, PTR1 has been the focus of recent research efforts aimed at finding more effective treatments against human parasitic diseases such as leishmaniasis or sleeping sickness. Previous PTR1-centered structural studies highlighted the enzyme characteristics, such as flexible regions around the active site, highly conserved structural waters, and species-specific differences in pocket properties and dynamics, which likely impacts the binding of natural substrates and inhibitors. Furthermore, several aspects of the PTR1 function, such as the substrate inhibition phenomenon and the level of ligand binding cooperativity in the enzyme homotetramer, likely related to the global enzyme dynamics, are poorly known at the molecular level. We postulate that future drug design efforts could greatly benefit from a better understanding of these phenomena through studying both the local and global PTR1 dynamics. This review highlights the key aspects of the PTR1 structure and dynamics relevant to structure-based drug design that could be effectively investigated by modeling approaches. Particular emphasis is given to the perspective of molecular dynamics, what has been accomplished in this area to date, and how modeling could impact the PTR1-targeted drug design in the future.
Collapse
Affiliation(s)
- Joanna Panecka-Hofman
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland.
| | - Ina Poehner
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211, Kuopio, Finland
| |
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
|
Rezaei AR, Saberi S, Sepehri S. Synthesis, Antileishmanial Activity and Molecular Docking Study of a Series of Dihydropyridine Derivatives. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2092877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Ali-Reza Rezaei
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Sedigheh Saberi
- Department of Mycology and Parasitology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Saghi Sepehri
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
5
|
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: 13] [Impact Index Per Article: 4.3] [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
|
6
|
Synthesis and Biological Evaluation of Tetrahydropyrimidine and Dihydropyridine Derivatives Against Leishmania Major. Acta Parasitol 2022; 67:255-266. [PMID: 34279776 DOI: 10.1007/s11686-021-00457-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE A number of tetrahydropyrimidines and their bioisosteric dihydropyridines bearing chloro substituent at various positions of phenyl ring in C4 of main scaffolds were designed, synthesized and evaluated for antileishmanial activity. METHODS The antileishmanial activity of the synthesized compounds was evaluated against promastigote and amastigote forms. Moreover, molecular docking studies of the compounds in pteridine reductase 1 (PTR1) pocket were carried out to describe the results of biological experiments. RESULTS The compounds exhibited moderate to good antileishmanial activity against promastigote and amastigote forms. Among the screened compounds, 1d and 2c were found as the most potent compounds against promastigote form with EC50 values of 15.5 and 10.5 µM, respectively. Compounds 2a and 2c were the most potent compounds against amastigote form with EC50 values of 5.4 and 2.2 µM, respectively. CONCLUSION According to structure-activity relationship (SAR) studies, the chloro substituent in different positions of phenyl ring at C4 of 1,2,3,4-tetrahydropyrimidine (THPM) and 1,4-dihydropyridine (DHP) rings and also the length of the chain belonging to the ester groups could be important for antileishmanial activity of these compounds. Most of these compounds exhibited low cytotoxicity against macrophages. Compounds 1 h, 2a, 2b and 2c revealed higher activity than glucantime while all compounds showed lower activity toward amphotericine B. Docking studies showed that the synthesized compounds were fit well in the PTR1 pocket. Compounds 1 h and 2c indicated the highest score docking among screened compounds in PTR1 enzyme.
Collapse
|
7
|
Ghanbariasad A, Emami L, Zarenezhad E, Behrouz S, Zarenezhad A, Soltani Rad MN. Synthesis, Biological Evaluation and In silico Studies of 1, 2, 3-triazolyl- Metronidazole Derivatives Against Leishmania Major. NEW J CHEM 2022. [DOI: 10.1039/d2nj00226d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The simple and effective approach for the preparation, of 1,2,3-triazolyl-based metronidazole hybrid analogues as promising anti-leishmania agents using of [CuL-SiO-HA] as a catalyst were described. The catalyst was fully characterized...
Collapse
|
8
|
Alcazar W, Alakurtti S, Padrón-Nieves M, Tuononen ML, Rodríguez N, Yli-Kauhaluoma J, Ponte-Sucre A. Leishmanicidal Activity of Betulin Derivatives in Leishmania amazonensis; Effect on Plasma and Mitochondrial Membrane Potential, and Macrophage Nitric Oxide and Superoxide Production. Microorganisms 2021; 9:320. [PMID: 33557150 PMCID: PMC7913927 DOI: 10.3390/microorganisms9020320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 11/25/2022] Open
Abstract
Herein, we evaluated in vitro the anti-leishmanial activity of betulin derivatives in Venezuelan isolates of Leishmania amazonensis, isolated from patients with therapeutic failure. METHODS We analyzed promastigote in vitro susceptibility as well as the cytotoxicity and selectivity of the evaluated compounds. Additionally, the activity of selected compounds was determined in intracellular amastigotes. Finally, to gain hints on their potential mechanism of action, the effect of the most promising compounds on plasma and mitochondrial membrane potential, and nitric oxide and superoxide production by infected macrophages was determined. RESULTS From the tested 28 compounds, those numbered 18 and 22 were chosen for additional studies. Both 18 and 22 were active (GI50 ≤ 2 µM, cytotoxic CC50 > 45 µM, SI > 20) for the reference strain LTB0016 and for patient isolates. The results suggest that 18 significantly depolarized the plasma membrane potential (p < 0.05) and the mitochondrial membrane potential (p < 0.05) when compared to untreated cells. Although neither 18 nor 22 induced nitric oxide production in infected macrophages, 18 induced superoxide production in infected macrophages. CONCLUSION Our results suggest that due to their efficacy and selectivity against intracellular parasites and the potential mechanisms underlying their leishmanicidal effect, the compounds 18 and 22 could be used as tools for designing new chemotherapies against leishmaniasis.
Collapse
Affiliation(s)
- Wilmer Alcazar
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, P.O. Box 50587, Caracas 1050, Venezuela; (W.A.); (M.P.-N.)
| | - Sami Alakurtti
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, P.O. Box 56, FI-00014 Helsinki, Finland; (S.A.); (M.L.T.)
- VTT Technical Research Centre of Finland, Biologinkuja 7, P.O. Box 1000, FI-02044 Espoo, Finland
| | - Maritza Padrón-Nieves
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, P.O. Box 50587, Caracas 1050, Venezuela; (W.A.); (M.P.-N.)
| | - Maija Liisa Tuononen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, P.O. Box 56, FI-00014 Helsinki, Finland; (S.A.); (M.L.T.)
- VTT Technical Research Centre of Finland, Biologinkuja 7, P.O. Box 1000, FI-02044 Espoo, Finland
| | - Noris Rodríguez
- Laboratory of Genetic Engineering, Institute of Biomedicine, Universidad Central de Venezuela, P.O. Box 4043, Caracas 1010A, Venezuela;
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, P.O. Box 56, FI-00014 Helsinki, Finland; (S.A.); (M.L.T.)
| | - Alicia Ponte-Sucre
- Laboratory of Molecular Physiology, Institute of Experimental Medicine, School of Medicine Luis Razetti, Faculty of Medicine, Universidad Central de Venezuela, P.O. Box 50587, Caracas 1050, Venezuela; (W.A.); (M.P.-N.)
| |
Collapse
|
9
|
Design, Synthesis and Biological Evaluation of Arylpyridin-2-yl Guanidine Derivatives and Cyclic Mimetics as Novel MSK1 Inhibitors. An Application in an Asthma Model. Molecules 2021; 26:molecules26020391. [PMID: 33450992 PMCID: PMC7828447 DOI: 10.3390/molecules26020391] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 01/23/2023] Open
Abstract
Mitogen- and Stress-Activated Kinase 1 (MSK1) is a nuclear kinase, taking part in the activation pathway of the pro-inflammatory transcription factor NF-kB and is demonstrating a therapeutic target potential in inflammatory diseases such as asthma, psoriasis and atherosclerosis. To date, few MSK1 inhibitors were reported. In order to identify new MSK1 inhibitors, a screening of a library of low molecular weight compounds was performed, and the results highlighted the 6-phenylpyridin-2-yl guanidine (compound 1a, IC50~18 µM) as a starting hit for structure-activity relationship study. Derivatives, homologues and rigid mimetics of 1a were designed, and all synthesized compounds were evaluated for their inhibitory activity towards MSK1. Among them, the non-cytotoxic 2-aminobenzimidazole 49d was the most potent at inhibiting significantly: (i) MSK1 activity, (ii) the release of IL-6 in inflammatory conditions in vitro (IC50~2 µM) and (iii) the inflammatory cell recruitment to the airways in a mouse model of asthma.
Collapse
|
10
|
Shamshad H, Hafiz A, Althagafi II, Saeed M, Mirza AZ. Characterization of the Trypanosoma brucei Pteridine Reductase Active- Site using Computational Docking and Virtual Screening Techniques. Curr Comput Aided Drug Des 2020; 16:583-598. [DOI: 10.2174/1573409915666190827163327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/21/2019] [Accepted: 08/01/2019] [Indexed: 01/19/2023]
Abstract
Background:
Human African trypanosomiasis is a fatal disease prevalent in approximately
36 sub-Saharan countries. Emerging reports of drug resistance in Trypanosoma brucei are a serious
cause of concern as only limited drugs are available for the treatment of the disease. Pteridine reductase
is an enzyme of Trypanosoma brucei.
Methods:
It plays a critical role in the pterin metabolic pathway that is absolutely essential for its survival
in the human host. The success of finding a potent inhibitor in structure-based drug design lies
within the ability of computational tools to efficiently and accurately dock a ligand into the binding
cavity of the target protein. Here we report the computational characterization of Trypanosoma brucei
pteridine reductase (Tb-PR) active-site using twenty-four high-resolution co-crystal structures with various
drugs. Structurally, the Tb-PR active site can be grouped in two clusters; one with high Root Mean
Square Deviation (RMSD) of atomic positions and another with low RMSD of atomic positions. These
clusters provide fresh insight for rational drug design against Tb-PR. Henceforth, the effect of several
factors on docking accuracy, including ligand and protein flexibility were analyzed using Fred.
Results:
The online server was used to analyze the side chain flexibility and four proteins were selected
on the basis of results. The proteins were subjected to small-scale virtual screening using 85 compounds,
and statistics were calculated using Bedroc and roc curves. The enrichment factor was also calculated
for the proteins and scoring functions. The best scoring function was used to understand the ligand
protein interactions with top common compounds of four proteins. In addition, we made a 3D
structural comparison between the active site of Tb-PR and Leishmania major pteridine reductase (Lm-
PR). We described key structural differences between Tb-PR and Lm-PR that can be exploited for rational
drug design against these two human parasites.
Conclusion:
The results indicated that relying just on re-docking and cross-docking experiments for
virtual screening of libraries isn’t enough and results might be misleading. Hence it has been suggested
that small scale virtual screening should be performed prior to large scale screening.
Collapse
Affiliation(s)
- Hina Shamshad
- Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, University of Karachi, Karachi-75270, Pakistan
| | - Abdul Hafiz
- Department of Medical Parasitology, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ismail I. Althagafi
- Chemistry Department, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Maria Saeed
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi- 75270, Pakistan
| | - Agha Zeeshan Mirza
- Chemistry Department, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
11
|
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: 1.0] [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
|
12
|
Pereira CA, Sayé M, Reigada C, Silber AM, Labadie GR, Miranda MR, Valera-Vera E. Computational approaches for drug discovery against trypanosomatid-caused diseases. Parasitology 2020; 147:611-633. [PMID: 32046803 PMCID: PMC10317681 DOI: 10.1017/s0031182020000207] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
During three decades, only about 20 new drugs have been developed for malaria, tuberculosis and all neglected tropical diseases (NTDs). This critical situation was reached because NTDs represent only 10% of health research investments; however, they comprise about 90% of the global disease burden. Computational simulations applied in virtual screening (VS) strategies are very efficient tools to identify pharmacologically active compounds or new indications for drugs already administered for other diseases. One of the advantages of this approach is the low time-consuming and low-budget first stage, which filters for testing experimentally a group of candidate compounds with high chances of binding to the target and present trypanocidal activity. In this work, we review the most common VS strategies that have been used for the identification of new drugs with special emphasis on those applied to trypanosomiasis and leishmaniasis. Computational simulations based on the selected protein targets or their ligands are explained, including the method selection criteria, examples of successful VS campaigns applied to NTDs, a list of validated molecular targets for drug development and repositioned drugs for trypanosomatid-caused diseases. Thereby, here we present the state-of-the-art of VS and drug repurposing to conclude pointing out the future perspectives in the field.
Collapse
Affiliation(s)
- Claudio A. Pereira
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Melisa Sayé
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Chantal Reigada
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Ariel M. Silber
- Laboratory of Biochemistry of Tryps – LaBTryps, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Guillermo R. Labadie
- Instituto de Química Rosario (IQUIR-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
- Departamento de Química Orgánica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Mariana R. Miranda
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| | - Edward Valera-Vera
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Investigaciones Médicas A. Lanari, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Investigaciones Médicas, Laboratorio de Parasitología Molecular, Buenos Aires, Argentina
| |
Collapse
|
13
|
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: 1.6] [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
|
14
|
Overcoming imatinib resistance in chronic myelogenous leukemia cells using non-cytotoxic cell death modulators. Eur J Med Chem 2019; 185:111748. [PMID: 31648125 DOI: 10.1016/j.ejmech.2019.111748] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/27/2019] [Accepted: 09/27/2019] [Indexed: 11/21/2022]
Abstract
Recent studies examined the possibility to overcome imatinib resistance in chronic myeloid leukemia (CML) patients by combination therapy with peroxisome proliferator-activated receptor gamma (PPARγ) ligands. Pioglitazone, a full PPARγ agonist, improved the survival of patients by the gradual elimination of the residual CML stem cell pool. To evaluate the importance of the pharmacological profile of PPARγ agonists on the ability to circumvent resistance, the partial PPARγ agonist 4'-((2-propyl-1H-benzo[d]imidazol-1-yl)methyl)-[1,1'-biphenyl]-2-carboxylic acid, derived from telmisartan, and other related derivatives were investigated. The 4-substituted benzimidazole derivatives bearing a [1,1'-biphenyl]-2-carboxamide moiety sensitized K562-resistant cells to imatinib treatment. Especially the derivatives 18a-f, which did not activate PPARγ to more than 40% at 10 μM, retrieved the cytotoxicity of imatinib in these cells. The cell death modulating properties were higher than that of pioglitazone. It is of interest to note that all novel compounds were not cytotoxic neither on non-resistant nor on resistant cells. They exerted antitumor potency only in combination with imatinib.
Collapse
|
15
|
Abstract
Parasites elicit several physiological changes in their host to enhance transmission. Little is known about the functional association between parasitism and microbiota-provisioned resources typically dedicated to animal hosts and how these goods may be rerouted to optimize parasite development. This study is the first to identify a specific symbiont-generated metabolite that impacts insect vector competence by facilitating parasite establishment and, thus, eventual transmission. Specifically, we demonstrate that the tsetse fly obligate mutualist Wigglesworthia provisions folate (vitamin B9) that pathogenic African trypanosomes exploit in an effort to successfully establish an infection in the vector’s MG. This process is essential for the parasite to complete its life cycle and be transmitted to a new vertebrate host. Disrupting metabolic contributions provided by the microbiota of arthropod disease vectors may fuel future innovative control strategies while also offering minimal nontarget effects. Many symbionts supplement their host’s diet with essential nutrients. However, whether these nutrients also enhance parasitism is unknown. In this study, we investigated whether folate (vitamin B9) production by the tsetse fly (Glossina spp.) essential mutualist, Wigglesworthia, aids auxotrophic African trypanosomes in completing their life cycle within this obligate vector. We show that the expression of Wigglesworthia folate biosynthesis genes changes with the progression of trypanosome infection within tsetse. The disruption of Wigglesworthia folate production caused a reduction in the percentage of flies that housed midgut (MG) trypanosome infections. However, decreased folate did not prevent MG trypanosomes from migrating to and establishing an infection in the fly’s salivary glands, thus suggesting that nutrient requirements vary throughout the trypanosome life cycle. We further substantiated that trypanosomes rely on symbiont-generated folate by feeding this vitamin to Glossina brevipalpis, which exhibits low trypanosome vector competency and houses Wigglesworthia incapable of producing folate. Folate-supplemented G. brevipalpis flies were significantly more susceptible to trypanosome infection, further demonstrating that this vitamin facilitates parasite infection establishment. Our cumulative results provide evidence that Wigglesworthia provides a key metabolite (folate) that is “hijacked” by trypanosomes to enhance their infectivity, thus indirectly impacting tsetse species vector competency. Parasite dependence on symbiont-derived micronutrients, which likely also occurs in other arthropod vectors, represents a relationship that may be exploited to reduce disease transmission.
Collapse
|
16
|
Synthesis and biological evaluation of solubilized sulfonamide analogues of the phosphatidylinositol 3-kinase inhibitor ZSTK474. Bioorg Med Chem 2019; 27:1529-1545. [DOI: 10.1016/j.bmc.2019.02.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/17/2019] [Accepted: 02/24/2019] [Indexed: 02/07/2023]
|
17
|
Linciano P, Pozzi C, Iacono LD, di Pisa F, Landi G, Bonucci A, Gul S, Kuzikov M, Ellinger B, Witt G, Santarem N, Baptista C, Franco C, Moraes CB, Müller W, Wittig U, Luciani R, Sesenna A, Quotadamo A, Ferrari S, Pöhner I, Cordeiro-da-Silva A, Mangani S, Costantino L, Costi MP. Enhancement of Benzothiazoles as Pteridine Reductase-1 Inhibitors for the Treatment of Trypanosomatidic Infections. J Med Chem 2019; 62:3989-4012. [DOI: 10.1021/acs.jmedchem.8b02021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Pasquale Linciano
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Cecilia Pozzi
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Lucia dello Iacono
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Flavio di Pisa
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Giacomo Landi
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Alessio Bonucci
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Gesa Witt
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port, 22525 Hamburg, Germany
| | - Nuno Santarem
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Catarina Baptista
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Caio Franco
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisaem Energia e Materiais (CNPEM), 13083-100 Campinas, São Paulo, Brazil
| | - Carolina B. Moraes
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisaem Energia e Materiais (CNPEM), 13083-100 Campinas, São Paulo, Brazil
| | | | | | - Rosaria Luciani
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Antony Sesenna
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Antonio Quotadamo
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Stefania Ferrari
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | | | - Anabela Cordeiro-da-Silva
- Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto and Institute for Molecular and Cell Biology, 4150-180 Porto, Portugal
| | - Stefano Mangani
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Luca Costantino
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Maria Paola Costi
- Dipartimento di Scienze della Vita, University of Modena and Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| |
Collapse
|
18
|
Jacobs L, de Kock C, Taylor D, Pelly SC, Blackie MA. Synthesis of five libraries of 6,5-fused heterocycles to establish the importance of the heterocyclic core for antiplasmodial activity. Bioorg Med Chem 2018; 26:5730-5741. [DOI: 10.1016/j.bmc.2018.10.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 09/24/2018] [Accepted: 10/26/2018] [Indexed: 10/28/2022]
|
19
|
Webster LA, Thomas M, Urbaniak M, Wyllie S, Ong H, Tinti M, Fairlamb AH, Boesche M, Ghidelli-Disse S, Drewes G, Gilbert IH. Development of Chemical Proteomics for the Folateome and Analysis of the Kinetoplastid Folateome. ACS Infect Dis 2018; 4:1475-1486. [PMID: 30264983 PMCID: PMC6199744 DOI: 10.1021/acsinfecdis.8b00097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
The folate pathway has been extensively
studied in a number of organisms, with its essentiality exploited
by a number of drugs. However, there has been little success in developing
drugs that target folate metabolism in the kinetoplastids. Despite
compounds being identified which show significant inhibition of the
parasite enzymes, this activity does not translate well into cellular
and animal models of disease. Understanding to which enzymes antifolates
bind under physiological conditions and how this corresponds to the
phenotypic response could provide insight on how to target the folate
pathway in these organisms. To facilitate this, we have adopted a
chemical proteomics approach to study binding of compounds to enzymes
of folate metabolism. Clinical and literature antifolate compounds
were immobilized onto resins to allow for “pull down”
of the proteins in the “folateome”. Using competition
studies, proteins, which bind the beads specifically and nonspecifically,
were identified in parasite lysate (Trypanosoma brucei and Leishmania major) for each antifolate compound.
Proteins were identified through tryptic digest, tandem mass tag (TMT)
labeling of peptides followed by LC-MS/MS. This approach was further
exploited by creating a combined folate resin (folate beads). The
resin could pull down up to 9 proteins from the folateome. This information
could be exploited in gaining a better understanding of folate metabolism
in kinetoplastids and other organisms.
Collapse
Affiliation(s)
- Lauren A. Webster
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Michael Thomas
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Michael Urbaniak
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Susan Wyllie
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Han Ong
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Michele Tinti
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Alan H. Fairlamb
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Markus Boesche
- Cellzome - a GSK company, Meyerhofstrasse 1, Heidelberg, 69117, Germany
| | | | - Gerard Drewes
- Cellzome - a GSK company, Meyerhofstrasse 1, Heidelberg, 69117, Germany
| | - Ian H. Gilbert
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| |
Collapse
|
20
|
Cullia G, Tamborini L, Conti P, De Micheli C, Pinto A. Folates in Trypanosoma brucei
: Achievements and Opportunities. ChemMedChem 2018; 13:2150-2158. [DOI: 10.1002/cmdc.201800500] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Gregorio Cullia
- Institute of Biomolecules Max Mousseron (IBMM); UMR5247; CNRS; University of Montpellier; ENSCM; Place Eugène Battaillon 34095 Montpellier cedex 5 France
| | - Lucia Tamborini
- Department of Pharmaceutical Sciences (DISFARM); University of Milan; via Luigi Mangiagalli 25 20133 Milano Italy
| | - Paola Conti
- Department of Pharmaceutical Sciences (DISFARM); University of Milan; via Luigi Mangiagalli 25 20133 Milano Italy
| | - Carlo De Micheli
- Department of Pharmaceutical Sciences (DISFARM); University of Milan; via Luigi Mangiagalli 25 20133 Milano Italy
| | - Andrea Pinto
- Department of Food Environmental and Nutritional Sciences; University of Milan; via Giovanni Celoria 2 20133 Milano Italy
| |
Collapse
|
21
|
Moreau RJ, Skepper CK, Appleton BA, Blechschmidt A, Balibar CJ, Benton BM, Drumm JE, Feng BY, Geng M, Li C, Lindvall MK, Lingel A, Lu Y, Mamo M, Mergo W, Polyakov V, Smith TM, Takeoka K, Uehara K, Wang L, Wei JR, Weiss AH, Xie L, Xu W, Zhang Q, de Vicente J. Fragment-Based Drug Discovery of Inhibitors of Phosphopantetheine Adenylyltransferase from Gram-Negative Bacteria. J Med Chem 2018; 61:3309-3324. [DOI: 10.1021/acs.jmedchem.7b01691] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Robert J. Moreau
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Colin K. Skepper
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Brent A. Appleton
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Anke Blechschmidt
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Carl J. Balibar
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Bret M. Benton
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Joseph E. Drumm
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Brian Y. Feng
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Mei Geng
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Cindy Li
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Mika K. Lindvall
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Andreas Lingel
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Yipin Lu
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Mulugeta Mamo
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Wosenu Mergo
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Valery Polyakov
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Thomas M. Smith
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Kenneth Takeoka
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Kyoko Uehara
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Lisha Wang
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Jun-Rong Wei
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Andrew H. Weiss
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Lili Xie
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Wenjian Xu
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Qiong Zhang
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Javier de Vicente
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| |
Collapse
|
22
|
Panecka-Hofman J, Pöhner I, Spyrakis F, Zeppelin T, Di Pisa F, Dello Iacono L, Bonucci A, Quotadamo A, Venturelli A, Mangani S, Costi M, Wade RC. Comparative mapping of on-targets and off-targets for the discovery of anti-trypanosomatid folate pathway inhibitors. Biochim Biophys Acta Gen Subj 2017; 1861:3215-3230. [DOI: 10.1016/j.bbagen.2017.09.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 01/06/2023]
|
23
|
Linciano P, Dawson A, Pöhner I, Costa DM, Sá MS, Cordeiro-da-Silva A, Luciani R, Gul S, Witt G, Ellinger B, Kuzikov M, Gribbon P, Reinshagen J, Wolf M, Behrens B, Hannaert V, Michels PAM, Nerini E, Pozzi C, di Pisa F, Landi G, Santarem N, Ferrari S, Saxena P, Lazzari S, Cannazza G, Freitas-Junior LH, Moraes CB, Pascoalino BS, Alcântara LM, Bertolacini CP, Fontana V, Wittig U, Müller W, Wade RC, Hunter WN, Mangani S, Costantino L, Costi MP. Exploiting the 2-Amino-1,3,4-thiadiazole Scaffold To Inhibit Trypanosoma brucei Pteridine Reductase in Support of Early-Stage Drug Discovery. ACS OMEGA 2017; 2:5666-5683. [PMID: 28983525 PMCID: PMC5623949 DOI: 10.1021/acsomega.7b00473] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/11/2017] [Indexed: 06/07/2023]
Abstract
Pteridine reductase-1 (PTR1) is a promising drug target for the treatment of trypanosomiasis. We investigated the potential of a previously identified class of thiadiazole inhibitors of Leishmania major PTR1 for activity against Trypanosoma brucei (Tb). We solved crystal structures of several TbPTR1-inhibitor complexes to guide the structure-based design of new thiadiazole derivatives. Subsequent synthesis and enzyme- and cell-based assays confirm new, mid-micromolar inhibitors of TbPTR1 with low toxicity. In particular, compound 4m, a biphenyl-thiadiazole-2,5-diamine with IC50 = 16 μM, was able to potentiate the antitrypanosomal activity of the dihydrofolate reductase inhibitor methotrexate (MTX) with a 4.1-fold decrease of the EC50 value. In addition, the antiparasitic activity of the combination of 4m and MTX was reversed by addition of folic acid. By adopting an efficient hit discovery platform, we demonstrate, using the 2-amino-1,3,4-thiadiazole scaffold, how a promising tool for the development of anti-T. brucei agents can be obtained.
Collapse
Affiliation(s)
- Pasquale Linciano
- Dipartimento di
Scienze della Vita, Università degli
Studi di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Alice Dawson
- Biological Chemistry &
Drug Discovery, School of Life Sciences, The Wellcome Trust Building, University of Dundee, Dow Street, Dundee DD1
5EH, U.K.
| | - Ina Pöhner
- Molecular
and Cellular Modeling Group and Scientific Databases and Visualization
(SDBV) Group, Heidelberg Institute for Theoretical
Studies, Schloss-Wolfsbrunnenweg
35, D-69118 Heidelberg, Germany
| | - David M. Costa
- Instituto de Investigação
e Inovação em Saúde, Instituto de Biologia Molecular
e Celular, and Departamento de Ciências Biológicas, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Monica S. Sá
- Instituto de Investigação
e Inovação em Saúde, Instituto de Biologia Molecular
e Celular, and Departamento de Ciências Biológicas, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Anabela Cordeiro-da-Silva
- Instituto de Investigação
e Inovação em Saúde, Instituto de Biologia Molecular
e Celular, and Departamento de Ciências Biológicas, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Rosaria Luciani
- Dipartimento di
Scienze della Vita, Università degli
Studi di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Sheraz Gul
- Fraunhofer-IME SP, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Gesa Witt
- Fraunhofer-IME SP, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | | | - Maria Kuzikov
- Fraunhofer-IME SP, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Philip Gribbon
- Fraunhofer-IME SP, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | | | - Markus Wolf
- Fraunhofer-IME SP, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Birte Behrens
- Fraunhofer-IME SP, Schnackenburgallee 114, D-22525 Hamburg, Germany
| | - Véronique Hannaert
- Research Unit for Tropical
Diseases, de Duve Institute and Laboratory of Biochemistry, Université catholique de Louvain, Avenue Hippocrate 74, B-1200 Brussels, Belgium
| | - Paul A. M. Michels
- Research Unit for Tropical
Diseases, de Duve Institute and Laboratory of Biochemistry, Université catholique de Louvain, Avenue Hippocrate 74, B-1200 Brussels, Belgium
| | - Erika Nerini
- Dipartimento di
Scienze della Vita, Università degli
Studi di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Cecilia Pozzi
- University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Flavio di Pisa
- University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Giacomo Landi
- University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Nuno Santarem
- Instituto de Investigação
e Inovação em Saúde, Instituto de Biologia Molecular
e Celular, and Departamento de Ciências Biológicas, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Stefania Ferrari
- Dipartimento di
Scienze della Vita, Università degli
Studi di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Puneet Saxena
- Dipartimento di
Scienze della Vita, Università degli
Studi di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Sandra Lazzari
- Dipartimento di
Scienze della Vita, Università degli
Studi di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Giuseppe Cannazza
- Dipartimento di
Scienze della Vita, Università degli
Studi di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Lucio H. Freitas-Junior
- Laboratório Nacional de Biociências CNPEM,
Centro Nacional de Pesquisa em Energia e Materials, Rua Giuseppe Máximo Scolfaro, 10.000, CEP 13083-970 Campinas/SP, Brasil
| | - Carolina B. Moraes
- Laboratório Nacional de Biociências CNPEM,
Centro Nacional de Pesquisa em Energia e Materials, Rua Giuseppe Máximo Scolfaro, 10.000, CEP 13083-970 Campinas/SP, Brasil
| | - Bruno S. Pascoalino
- Laboratório Nacional de Biociências CNPEM,
Centro Nacional de Pesquisa em Energia e Materials, Rua Giuseppe Máximo Scolfaro, 10.000, CEP 13083-970 Campinas/SP, Brasil
| | - Laura M. Alcântara
- Laboratório Nacional de Biociências CNPEM,
Centro Nacional de Pesquisa em Energia e Materials, Rua Giuseppe Máximo Scolfaro, 10.000, CEP 13083-970 Campinas/SP, Brasil
| | - Claudia P. Bertolacini
- Laboratório Nacional de Biociências CNPEM,
Centro Nacional de Pesquisa em Energia e Materials, Rua Giuseppe Máximo Scolfaro, 10.000, CEP 13083-970 Campinas/SP, Brasil
| | - Vanessa Fontana
- Laboratório Nacional de Biociências CNPEM,
Centro Nacional de Pesquisa em Energia e Materials, Rua Giuseppe Máximo Scolfaro, 10.000, CEP 13083-970 Campinas/SP, Brasil
| | - Ulrike Wittig
- Molecular
and Cellular Modeling Group and Scientific Databases and Visualization
(SDBV) Group, Heidelberg Institute for Theoretical
Studies, Schloss-Wolfsbrunnenweg
35, D-69118 Heidelberg, Germany
| | - Wolfgang Müller
- Molecular
and Cellular Modeling Group and Scientific Databases and Visualization
(SDBV) Group, Heidelberg Institute for Theoretical
Studies, Schloss-Wolfsbrunnenweg
35, D-69118 Heidelberg, Germany
| | - Rebecca C. Wade
- Molecular
and Cellular Modeling Group and Scientific Databases and Visualization
(SDBV) Group, Heidelberg Institute for Theoretical
Studies, Schloss-Wolfsbrunnenweg
35, D-69118 Heidelberg, Germany
- Center for Molecular Biology (ZMBH), DKFZ−ZMBH Alliance, Heidelberg University, Im Neuenheimer Feld 282, D-69120 Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Im Neuenheimer Feld 205, D-69120 Heidelberg, Germany
| | - William N. Hunter
- Biological Chemistry &
Drug Discovery, School of Life Sciences, The Wellcome Trust Building, University of Dundee, Dow Street, Dundee DD1
5EH, U.K.
| | | | - Luca Costantino
- Dipartimento di
Scienze della Vita, Università degli
Studi di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Maria P. Costi
- Dipartimento di
Scienze della Vita, Università degli
Studi di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| |
Collapse
|
24
|
Jedwabny W, Panecka-Hofman J, Dyguda-Kazimierowicz E, Wade RC, Sokalski WA. Application of a simple quantum chemical approach to ligand fragment scoring for Trypanosoma brucei pteridine reductase 1 inhibition. J Comput Aided Mol Des 2017; 31:715-728. [PMID: 28688090 PMCID: PMC5570812 DOI: 10.1007/s10822-017-0035-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 06/16/2017] [Indexed: 11/15/2022]
Abstract
There is a need for improved and generally applicable scoring functions for fragment-based approaches to ligand design. Here, we evaluate the performance of a computationally efficient model for inhibitory activity estimation, which is composed only of multipole electrostatic energy and dispersion energy terms that approximate long-range ab initio quantum mechanical interaction energies. We find that computed energies correlate well with inhibitory activity for a compound series with varying substituents targeting two subpockets of the binding site of Trypanosoma brucei pteridine reductase 1. For one subpocket, we find that the model is more predictive for inhibitory activity than the ab initio interaction energy calculated at the MP2 level. Furthermore, the model is found to outperform a commonly used empirical scoring method. Finally, we show that the results for the two subpockets can be combined, which suggests that this simple nonempirical scoring function could be applied in fragment–based drug design.
Collapse
Affiliation(s)
- Wiktoria Jedwabny
- Department of Chemistry, Wrocław University of Science and Technology, Wrocław, Poland
| | - Joanna Panecka-Hofman
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany.,Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | | | - Rebecca C Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany.,Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany.,Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Heidelberg, Germany
| | - W Andrzej Sokalski
- Department of Chemistry, Wrocław University of Science and Technology, Wrocław, Poland
| |
Collapse
|
25
|
Dewar S, Sienkiewicz N, Ong HB, Wall RJ, Horn D, Fairlamb AH. The Role of Folate Transport in Antifolate Drug Action in Trypanosoma brucei. J Biol Chem 2016; 291:24768-24778. [PMID: 27703008 PMCID: PMC5114424 DOI: 10.1074/jbc.m116.750422] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/14/2016] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to identify and characterize mechanisms of resistance to antifolate drugs in African trypanosomes. Genome-wide RNAi library screens were undertaken in bloodstream form Trypanosoma brucei exposed to the antifolates methotrexate and raltitrexed. In conjunction with drug susceptibility and folate transport studies, RNAi knockdown was used to validate the functions of the putative folate transporters. The transport kinetics of folate and methotrexate were further characterized in whole cells. RNA interference target sequencing experiments identified a tandem array of genes encoding a folate transporter family, TbFT1-3, as major contributors to antifolate drug uptake. RNAi knockdown of TbFT1-3 substantially reduced folate transport into trypanosomes and reduced the parasite's susceptibly to the classical antifolates methotrexate and raltitrexed. In contrast, knockdown of TbFT1-3 increased susceptibly to the non-classical antifolates pyrimethamine and nolatrexed. Both folate and methotrexate transport were inhibited by classical antifolates but not by non-classical antifolates or biopterin. Thus, TbFT1-3 mediates the uptake of folate and classical antifolates in trypanosomes, and TbFT1-3 loss-of-function is a mechanism of antifolate drug resistance.
Collapse
Affiliation(s)
- Simon Dewar
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Natasha Sienkiewicz
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Han B Ong
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Richard J Wall
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - David Horn
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Alan H Fairlamb
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom.
| |
Collapse
|
26
|
Mai S, Zhao Y, Song Q. Chemoselective acylation of benzimidazoles with phenylacetic acids under different Cu catalysts to give fused five-membered N-heterocycles or tertiary amides. Org Biomol Chem 2016; 14:8685-90. [PMID: 27430929 DOI: 10.1039/c6ob01167e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
C-N bond formation via a copper-catalyzed aerobic oxidative decarboxylative tandem protocol was realized. The phenylacetic acids which contain ortho-X (X = F or Br) on the aromatic ring will render a fused five-membered heterocycle via a tandem aromatic nucleophilic substitution and aerobic oxidative decarboxylative acylation at the C(sp(2))-H bond of benzimidazoles under the Cu(OAc)2/K2CO3/BF3·Et2O catalytic system, while with CuBr as the catalyst and pyridine as the base, N-acylation occurred and tertiary amides were obtained.
Collapse
Affiliation(s)
- Shaoyu Mai
- Institute of Next Generation Matter Transformation, College of Chemical Engineering at Huaqiao University, China.
| | | | | |
Collapse
|
27
|
Masoud GN, Wang J, Chen J, Miller D, Li W. Design, Synthesis and Biological Evaluation of Novel HIF1α Inhibitors. Anticancer Res 2015; 35:3849-3859. [PMID: 26124330 PMCID: PMC5915369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Elevated levels of hypoxia inducible factor-1 (HIF1) are linked to tumor metastasis, angiogenesis, poor patient prognosis and response to chemotherapy. HIF1α is a master regulator of the hypoxic response, including in cancer cells, through transcriptional activation of several target genes. Previously, we identified compound CJ-3k with high anti-HIF1α activity based on the structure of a well-known HIF1α inhibitor, YC-1. In this study, the CJ-3k scaffold was systematically modified to explore the structure-activity relationships. Fifty-three new CJ-3k analogs were synthesized and screened for their anti-HIF-1α activity in a luciferase-transfected human breast cancer cell line (MDA-MB-231). Some of these new analogs have a significantly greater activity than that of CJ-3k and hold potential for development as new therapeutic agents for the treatment of cancer.
Collapse
Affiliation(s)
- Georgina N Masoud
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Jin Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Jianjun Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, U.S.A. Department of Pharmaceutical Sciences, School of Pharmacy, South College, Knoxville, TN, U.S.A
| | - Duane Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, U.S.A.
| |
Collapse
|
28
|
Ding W, Song Q. Cu-catalyzed aerobic oxidative amidation of aryl alkyl ketones with azoles to afford tertiary amides via selective C–C bond cleavage. Org Chem Front 2015. [DOI: 10.1039/c5qo00101c] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Chemoselective cleavage of the C(CO)–C(alkyl) bond in aryl ketones leading to azole amides is disclosed. Aryl ketones with a variety of long-chain alkyl groups have been demonstrated to be active substrates and mechanism studies suggested that molecular oxygen serves both as an oxidant and a reactant in this strategy.
Collapse
Affiliation(s)
- Wen Ding
- Institute of Next Generation Matter Transformation
- College of Chemical Engineering at Huaqiao University
- Xiamen
- P. R. China
| | - Qiuling Song
- Institute of Next Generation Matter Transformation
- College of Chemical Engineering at Huaqiao University
- Xiamen
- P. R. China
- Beijing National Laboratory for Molecular Sciences
| |
Collapse
|
29
|
Keri RS, Hiremathad A, Budagumpi S, Nagaraja BM. Comprehensive Review in Current Developments of Benzimidazole-Based Medicinal Chemistry. Chem Biol Drug Des 2014; 86:19-65. [PMID: 25352112 DOI: 10.1111/cbdd.12462] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/12/2014] [Indexed: 12/13/2022]
Abstract
The properties of benzimidazole and its derivatives have been studied over more than one hundred years. Benzimidazole derivatives are useful intermediates/subunits for the development of molecules of pharmaceutical or biological interest. Substituted benzimidazole derivatives have found applications in diverse therapeutic areas such as antiulcer, anticancer agents, and anthelmintic species to name just a few. This work systematically gives a comprehensive review in current developments of benzimidazole-based compounds in the whole range of medicinal chemistry as anticancer, antibacterial, antifungal, anti-inflammatory, analgesic agents, anti-HIV, antioxidant, anticonvulsant, antitubercular, antidiabetic, antileishmanial, antihistaminic, antimalarial agents, and other medicinal agents. This review will further be helpful for the researcher on the basis of substitution pattern around the nucleus with an aim to help medicinal chemists for developing an SAR on benzimidazole drugs/compounds.
Collapse
Affiliation(s)
- Rangappa S Keri
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bangalore, Karnataka, 562112, India
| | - Asha Hiremathad
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bangalore, Karnataka, 562112, India
| | - Srinivasa Budagumpi
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bangalore, Karnataka, 562112, India
| | - Bhari Mallanna Nagaraja
- Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bangalore, Karnataka, 562112, India
| |
Collapse
|
30
|
Kryshchyshyn A, Kaminskyy D, Grellier P, Lesyk R. Trends in research of antitrypanosomal agents among synthetic heterocycles. Eur J Med Chem 2014; 85:51-64. [DOI: 10.1016/j.ejmech.2014.07.092] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 12/18/2022]
|
31
|
Khalaf A, Huggan JK, Suckling CJ, Gibson C, Stewart K, Giordani F, Barrett MP, Wong PE, Barrack KL, Hunter WN. Structure-based design and synthesis of antiparasitic pyrrolopyrimidines targeting pteridine reductase 1. J Med Chem 2014; 57:6479-94. [PMID: 25007262 PMCID: PMC4136963 DOI: 10.1021/jm500483b] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The treatment of Human African trypanosomiasis remains a major unmet health need in sub-Saharan Africa. Approaches involving new molecular targets are important; pteridine reductase 1 (PTR1), an enzyme that reduces dihydrobiopterin in Trypanosoma spp., has been identified as a candidate target, and it has been shown previously that substituted pyrrolo[2,3-d]pyrimidines are inhibitors of PTR1 from Trypanosoma brucei (J. Med. Chem. 2010, 53, 221-229). In this study, 61 new pyrrolo[2,3-d]pyrimidines have been prepared, designed with input from new crystal structures of 23 of these compounds complexed with PTR1, and evaluated in screens for enzyme inhibitory activity against PTR1 and in vitro antitrypanosomal activity. Eight compounds were sufficiently active in both screens to take forward to in vivo evaluation. Thus, although evidence for trypanocidal activity in a stage I disease model in mice was obtained, the compounds were too toxic to mice for further development.
Collapse
Affiliation(s)
- Abedawn
I. Khalaf
- WestCHEM
Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
| | - Judith K. Huggan
- WestCHEM
Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
| | - Colin J. Suckling
- WestCHEM
Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom, (C.J.S.) E-mail: ; Tel.: +44 141 548 2271
| | - Colin
L. Gibson
- WestCHEM
Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
| | - Kirsten Stewart
- WestCHEM
Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
| | - Federica Giordani
- Wellcome
Trust Centre for Molecular Parasitology, Institute of Infection, Immunity
and Inflammation and Glasgow Polyomics, College of Medical, Veterinary
and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Michael P. Barrett
- Wellcome
Trust Centre for Molecular Parasitology, Institute of Infection, Immunity
and Inflammation and Glasgow Polyomics, College of Medical, Veterinary
and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom,(M.P.B.) E-mail: ; Tel.: +44 141 330 6904
| | - Pui Ee Wong
- Wellcome
Trust Centre for Molecular Parasitology, Institute of Infection, Immunity
and Inflammation and Glasgow Polyomics, College of Medical, Veterinary
and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Keri L. Barrack
- Division
of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, United
Kingdom
| | - William N. Hunter
- Division
of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, DD1 5EH, United
Kingdom,(W.N.H.) E-mail: ; Tel.: +44 1382 385745
| |
Collapse
|
32
|
Drug discovery and human African trypanosomiasis: a disease less neglected? Future Med Chem 2014; 5:1801-41. [PMID: 24144414 DOI: 10.4155/fmc.13.162] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Human African trypanosomiasis (HAT) has been neglected for a long time. The most recent drug to treat this disease, eflornithine, was approved by the US FDA in 2000. Current treatments exhibit numerous problematic side effects and are often ineffective against the debilitating CNS resident stage of the disease. Fortunately, several partnerships and initiatives have been formed over the last 20 years in an effort to eradicate HAT, along with a number of other neglected diseases. This has led to an increasing number of foundations and research institutions that are currently working on the development of new drugs for HAT and tools with which to diagnose and treat patients. New biochemical pathways as therapeutic targets are emerging, accompanied by increasing numbers of new antitrypanosomal compound classes. The future looks promising that this collaborative approach will facilitate eagerly awaited breakthroughs in the treatment of HAT.
Collapse
|
33
|
Dube D, Sharma S, Singh TP, Kaur P. Pharmacophore Mapping, In Silico Screening and Molecular Docking to Identify SelectiveTrypanosoma bruceiPteridine Reductase Inhibitors. Mol Inform 2014; 33:124-34. [DOI: 10.1002/minf.201300023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 11/05/2013] [Indexed: 12/17/2022]
|
34
|
Gilbert IH. Drug discovery for neglected diseases: molecular target-based and phenotypic approaches. J Med Chem 2013; 56:7719-26. [PMID: 24015767 PMCID: PMC3954685 DOI: 10.1021/jm400362b] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
![]()
Drug
discovery for neglected tropical diseases is carried out using
both target-based and phenotypic approaches. In this paper, target-based
approaches are discussed, with a particular focus on human African
trypanosomiasis. Target-based drug discovery can be successful, but
careful selection of targets is required. There are still very few
fully validated drug targets in neglected diseases, and there is a
high attrition rate in target-based drug discovery for these diseases.
Phenotypic screening is a powerful method in both neglected and non-neglected
diseases and has been very successfully used. Identification of molecular
targets from phenotypic approaches can be a way to identify potential
new drug targets.
Collapse
Affiliation(s)
- Ian H Gilbert
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee , Dundee DD1 5EH, U.K
| |
Collapse
|
35
|
Woodland A, Grimaldi R, Luksch T, Cleghorn LAT, Ojo KK, Van Voorhis WC, Brenk R, Frearson JA, Gilbert IH, Wyatt PG. From on-target to off-target activity: identification and optimisation of Trypanosoma brucei GSK3 inhibitors and their characterisation as anti-Trypanosoma brucei drug discovery lead molecules. ChemMedChem 2013; 8:1127-37. [PMID: 23776181 PMCID: PMC3728731 DOI: 10.1002/cmdc.201300072] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 05/15/2013] [Indexed: 01/09/2023]
Abstract
Human African trypanosomiasis (HAT) is a life-threatening disease with approximately 30 000–40 000 new cases each year. Trypanosoma brucei protein kinase GSK3 short (TbGSK3) is required for parasite growth and survival. Herein we report a screen of a focused kinase library against T. brucei GSK3. From this we identified a series of several highly ligand-efficient TbGSK3 inhibitors. Following the hit validation process, we optimised a series of diaminothiazoles, identifying low-nanomolar inhibitors of TbGSK3 that are potent in vitro inhibitors of T. brucei proliferation. We show that the TbGSK3 pharmacophore overlaps with that of one or more additional molecular targets.
Collapse
Affiliation(s)
- Andrew Woodland
- Drug Discovery Unit (DDU), Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Sir James Black Centre, DD1 5EH, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Di Braccio M, Grossi G, Signorello MG, Leoncini G, Cichero E, Fossa P, Alfei S, Damonte G. Synthesis, in vitro antiplatelet activity and molecular modelling studies of 10-substituted 2-(1-piperazinyl)pyrimido[1,2-a]benzimidazol-4(10H)-ones. Eur J Med Chem 2013; 62:564-78. [PMID: 23425969 DOI: 10.1016/j.ejmech.2013.01.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 12/12/2012] [Accepted: 01/21/2013] [Indexed: 11/26/2022]
Abstract
The multistep preparation of the new 10-substituted 2-(1-piperazinyl)pyrimido[1,2-a]benzimidazol-4(10H)-ones 6a-o, and of the two isomers 10-ethyl-2-(diethylamino)pyrimido[1,2-a]benzimidazol-4(10H)-one 6p and 10-ethyl-4-(diethylamino)pyrimido[1,2-a]benzimidazol-2(10H)-one 13, as well as the in vitro evaluation of their inhibitory activity on human platelet aggregation induced in platelet-rich plasma by ADP, collagen or the Ca(2+) ionophore A23187 were here described. Nine out of fifteen 2-(1-piperazinyl)derivatives (6g-o) showed good inhibitory properties towards all the platelet aggregation agonists used. Moreover, a molecular modelling study has been performed on two of the best compounds of this series (6i and 6o) to confirm in silico their interactions with the catalytic site of human platelet PDE3, using the X-ray data of the PDE3B isoform in complex with an inhibitor.
Collapse
Affiliation(s)
- Mario Di Braccio
- Dipartimento di Farmacia, Sezione Chimica Farmaceutica, Università di Genova, Viale Benedetto XV, 16132 Genoa, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
I. H. Gilbert. ChemMedChem 2011. [DOI: 10.1002/cmdc.201100414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
38
|
Dube D, Periwal V, Kumar M, Sharma S, Singh TP, Kaur P. 3D-QSAR based pharmacophore modeling and virtual screening for identification of novel pteridine reductase inhibitors. J Mol Model 2011; 18:1701-11. [PMID: 21826447 DOI: 10.1007/s00894-011-1187-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 07/12/2011] [Indexed: 10/17/2022]
Abstract
Pteridine reductase is a promising target for development of novel therapeutic agents against Trypanosomatid parasites. A 3D-QSAR pharmacophore hypothesis has been generated for a series of L. major pteridine reductase inhibitors using Catalyst/HypoGen algorithm for identification of the chemical features that are responsible for the inhibitory activity. Four pharmacophore features, namely: two H-bond donors (D), one Hydrophobic aromatic (H) and one Ring aromatic (R) have been identified as key features involved in inhibitor-PTR1 interaction. These features are able to predict the activity of external test set of pteridine reductase inhibitors with a correlation coefficient (r) of 0.80. Based on the analysis of the best hypotheses, some potent Pteridine reductase inhibitors were screened out and predicted with anti-PTR1 activity. It turned out that the newly identified inhibitory molecules are at least 300 fold more potent than the current crop of existing inhibitors. Overall the current SAR study is an effort for elucidating quantitative structure-activity relationship for the PTR1 inhibitors. The results from the combined 3D-QSAR modeling and molecular docking approach have led to the prediction of new potent inhibitory scaffolds.
Collapse
Affiliation(s)
- Divya Dube
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | | | | | | | | |
Collapse
|