1
|
Sharma M, Lolli ML, Vyas VK. A comprehensive review of synthetic strategies and SAR studies for the discovery of PfDHODH inhibitors as antimalarial agents. Part 2: Non-DSM compounds. Bioorg Chem 2024; 153:107754. [PMID: 39241585 DOI: 10.1016/j.bioorg.2024.107754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024]
Abstract
Malaria remains a severe global health concern, with 249 million cases reported in 2022, according to the World Health Organization (WHO) [1]. PfDHODH is an essential enzyme in malaria parasites that helps to synthesize certain building blocks for their growth and development. It has been confirmed that targeting Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) enzyme could lead to new and effective antimalarial drugs. Inhibitors of PfDHODH have shown potential for slowing down parasite growth during both the blood and liver stages. Over the last two decades, many species selective PfDHODH inhibitors have been designed, including DSM compounds and other non-DSM compounds. In the first chapter [2] of this review, we have reviewed all synthetic schemes and structure-activity relationship (SAR) studies of DSM compounds. In this second chapter, we have compiled all the other non-DSM PfDHODH inhibitors based on dihydrothiophenones, thiazoles, hydroxyazoles, and N-alkyl-thiophene-2-carboxamides. The review not only offers an insightful overview of the synthetic methods employed but also explores into alternative routes and innovative strategies involving different catalysts and chemical reagents. A critical aspect covered in the review is the SAR studies, which provide a comprehensive understanding of how structural modifications impact the efficacy of PfDHODH inhibitors and challenges related to the discovery of PfDHODH inhibitors. This information is invaluable for scientists engaged in the development of new antimalarial drugs, offering insights into the most promising scaffolds and their synthetic techniques.
Collapse
Affiliation(s)
- Manmohan Sharma
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India
| | - Marco L Lolli
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India.
| |
Collapse
|
2
|
Sharma M, Pandey V, Poli G, Tuccinardi T, Lolli ML, Vyas VK. A comprehensive review of synthetic strategies and SAR studies for the discovery of PfDHODH inhibitors as antimalarial agents. Part 1: triazolopyrimidine, isoxazolopyrimidine and pyrrole-based (DSM) compounds. Bioorg Chem 2024; 146:107249. [PMID: 38493638 DOI: 10.1016/j.bioorg.2024.107249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/10/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024]
Abstract
One of the deadliest infectious diseases, malaria, still has a significant impact on global morbidity and mortality. Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) catalyzes the fourth step in de novo pyrimidine nucleotide biosynthesis and has been clinically validated as an innovative and promising target for the development of novel targeted antimalarial drugs. PfDHODH inhibitors have the potential to significantly slow down parasite growth at the blood and liver stages. Several PfDHODH inhibitors based on various scaffolds have been explored over the past two decades. Among them, triazolopyrimidines, isoxazolopyrimidines, and pyrrole-based derivatives known as DSM compounds showed tremendous potential as novel antimalarial agents, and one of the triazolopyrimidine-based compounds (DSM265) was able to reach phase IIa clinical trials. DSM compounds were synthesized as PfDHODH inhibitors with various substitutions based on structure-guided medicinal chemistry approaches and further optimised as well. For the first time, this review provides an overview of all the synthetic approaches used for the synthesis, alternative synthetic routes, and novel strategies involving various catalysts and chemical reagents that have been used to synthesize DSM compounds. We have also summarized SAR study of all these PfDHODH inhibitors. In an attempt to assist readers, scientists, and researchers involved in the development of new PfDHODH inhibitors as antimalarials, this review provides accessibility of all synthetic techniques and SAR studies of the most promising triazolopyrimidines, isoxazolopyrimidines, and pyrrole-based PfDHODH inhibitors.
Collapse
Affiliation(s)
- Manmohan Sharma
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India
| | - Vinita Pandey
- MIT College of Pharmacy, Ramganga Vihar, Phase-II, Moradabad, UP-244001, India
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Marco L Lolli
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 - Turin, Italy
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad 382481, India.
| |
Collapse
|
3
|
Vyas VK, Shukla T, Sharma M. Medicinal chemistry approaches for the discovery of Plasmodium falciparum dihydroorotate dehydrogenase inhibitors as antimalarial agents. Future Med Chem 2023; 15:1295-1321. [PMID: 37551689 DOI: 10.4155/fmc-2023-0113] [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: 08/09/2023] Open
Abstract
Malaria is a severe human disease and a global health problem because of drug-resistant strains. Drugs reported to prevent the growth of Plasmodium parasites target various phases of the parasites' life cycle. Antimalarial drugs can inhibit key enzymes that are responsible for the cellular growth and development of parasites. Plasmodium falciparum dihydroorotate dehydrogenase is one such enzyme that is necessary for de novo pyrimidine biosynthesis. This review focuses on various medicinal chemistry approaches used for the discovery and identification of selective P. falciparum dihydroorotate dehydrogenase inhibitors as antimalarial agents. This comprehensive review discusses recent advances in the selective therapeutic activity of distinct chemical classes of compounds as P. falciparum dihydroorotate dehydrogenase inhibitors and antimalarial drugs.
Collapse
Affiliation(s)
- Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Tanvi Shukla
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| | - Manmohan Sharma
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, 382481, India
| |
Collapse
|
4
|
Vah L, Medved T, Grošelj U, Klemenčič M, Podlipnik Č, Štefane B, Wagger J, Novinec M, Svete J. Regioselective Synthesis of 5- and 3-Hydroxy- N-Aryl-1 H-Pyrazole-4-Carboxylates and Their Evaluation as Inhibitors of Plasmodium falciparum Dihydroorotate Dehydrogenase. Molecules 2022; 27:4764. [PMID: 35897941 PMCID: PMC9332393 DOI: 10.3390/molecules27154764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 02/05/2023] Open
Abstract
In silico evaluation of various regioisomeric 5- and 3-hydroxy-substituted alkyl 1-aryl-1H-pyrazole-4-carboxylates and their acyclic precursors yielded promising results with respect to their binding in the active site of dihydroorotate dehydrogenase of Plasmodium falciparum (PfDHODH). Consequently, four ethyl 1-aryl-5-hydroxy-1H-pyrazole-4-carboxylates and their 3-hydroxy regioisomers were prepared by two-step syntheses via enaminone-type reagents or key intermediates. The synthesis of 5-hydroxy-1H-pyrazoles was carried out using the literature protocol comprising acid-catalyzed transamination of diethyl [(dimethylamino)methylene]malonate with arylhydrazines followed by base-catalyzed cyclization of the intermediate hydrazones. For the synthesis of isomeric methyl 1-aryl-3-hydroxy-1H-pyrazole-4-carboxylates, a novel two-step synthesis was developed. It comprises acylation of hydrazines with methyl malonyl chloride followed by cyclization of the hydrazines with tert-butoxy-bis(dimethylamino)methane. Testing the pyrazole derivatives for the inhibition of PfDHODH showed that 1-(naphthalene-2-yl)-5-hydroxy-1H-pyrazole-4-carboxylate and 1-(naphthalene-2-yl)-, 1-(2,4,6-trichlorophenyl)-, and 1-[4-(trifluoromethyl)phenyl]-3-hydroxy-1H-pyrazole-4-carboxylates (~30% inhibition) were slightly more potent than a known inhibitor, diethyl α-{[(1H-indazol-5-yl)amino]methylidene}malonate (19% inhibition).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Marko Novinec
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, 1000 Ljubljana, Slovenia; (L.V.); (T.M.); (U.G.); (M.K.); (Č.P.); (B.Š.); (J.W.)
| | - Jurij Svete
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, 1000 Ljubljana, Slovenia; (L.V.); (T.M.); (U.G.); (M.K.); (Č.P.); (B.Š.); (J.W.)
| |
Collapse
|
5
|
Yang T, Ottilie S, Istvan ES, Godinez-Macias KP, Lukens AK, Baragaña B, Campo B, Walpole C, Niles JC, Chibale K, Dechering KJ, Llinás M, Lee MCS, Kato N, Wyllie S, McNamara CW, Gamo FJ, Burrows J, Fidock DA, Goldberg DE, Gilbert IH, Wirth DF, Winzeler EA. MalDA, Accelerating Malaria Drug Discovery. Trends Parasitol 2021; 37:493-507. [PMID: 33648890 PMCID: PMC8261838 DOI: 10.1016/j.pt.2021.01.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/24/2022]
Abstract
The Malaria Drug Accelerator (MalDA) is a consortium of 15 leading scientific laboratories. The aim of MalDA is to improve and accelerate the early antimalarial drug discovery process by identifying new, essential, druggable targets. In addition, it seeks to produce early lead inhibitors that may be advanced into drug candidates suitable for preclinical development and subsequent clinical testing in humans. By sharing resources, including expertise, knowledge, materials, and reagents, the consortium strives to eliminate the structural barriers often encountered in the drug discovery process. Here we discuss the mission of the consortium and its scientific achievements, including the identification of new chemically and biologically validated targets, as well as future scientific directions.
Collapse
Affiliation(s)
- Tuo Yang
- Department of Pediatrics, School of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Sabine Ottilie
- Department of Pediatrics, School of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Eva S Istvan
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO 63130, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63130, USA
| | - Karla P Godinez-Macias
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Amanda K Lukens
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA 02142, USA
| | - Beatriz Baragaña
- Wellcome Center for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 5EH, UK
| | - Brice Campo
- Medicines for Malaria Venture, 1215 Geneva 15, Switzerland
| | - Chris Walpole
- Structural Genomics Consortium, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Jacquin C Niles
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Building 56-341, 77 Massachusetts Avenue, Cambridge MA 02139-4307, USA
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa; South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | | | - Manuel Llinás
- Department of Biochemistry and Molecular Biology and Department of Chemistry, Huck Center for Malaria Research, The Pennsylvania State University, University Park, PA 16082, USA
| | - Marcus C S Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Nobutaka Kato
- Global Health Drug Discovery Institute, Zhongguancun Dongsheng International Science Park, 1 North Yongtaizhuang Road, Beijing 100192, China
| | - Susan Wyllie
- Wellcome Center for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 5EH, UK
| | - Case W McNamara
- Calibr, a division of The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Francisco Javier Gamo
- Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, 28760, Madrid, Spain
| | - Jeremy Burrows
- Medicines for Malaria Venture, 1215 Geneva 15, Switzerland
| | - David A Fidock
- Department of Microbiology and Immunology and Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Daniel E Goldberg
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO 63130, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63130, USA
| | - Ian H Gilbert
- Wellcome Center for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee DD1 5EH, UK
| | - Dyann F Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA 02142, USA
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92093, USA.
| |
Collapse
|
6
|
Driving antimalarial design through understanding of target mechanism. Biochem Soc Trans 2020; 48:2067-2078. [PMID: 32869828 PMCID: PMC7609028 DOI: 10.1042/bst20200224] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 11/17/2022]
Abstract
Malaria continues to be a global health threat, affecting approximately 219 million people in 2018 alone. The recurrent development of resistance to existing antimalarials means that the design of new drug candidates must be carefully considered. Understanding of drug target mechanism can dramatically accelerate early-stage target-based development of novel antimalarials and allows for structural modifications even during late-stage preclinical development. Here, we have provided an overview of three promising antimalarial molecular targets, PfDHFR, PfDHODH and PfA-M1, and their associated inhibitors which demonstrate how mechanism can inform drug design and be effectively utilised to generate compounds with potent inhibitory activity.
Collapse
|
7
|
Madhav H, Hoda N. An insight into the recent development of the clinical candidates for the treatment of malaria and their target proteins. Eur J Med Chem 2020; 210:112955. [PMID: 33131885 DOI: 10.1016/j.ejmech.2020.112955] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 01/18/2023]
Abstract
Malaria is an endemic disease, prevalent in tropical and subtropical regions which cost half of million deaths annually. The eradication of malaria is one of the global health priority nevertheless, current therapeutic efforts seem to be insufficient due to the emergence of drug resistance towards most of the available drugs, even first-line treatment ACT, unavailability of the vaccine, and lack of drugs with a new mechanism of action. Intensification of antimalarial research in recent years has resulted into the development of single dose multistage therapeutic agents which has advantage of overcoming the antimalarial drug resistance. The present review explored the current progress in the development of new promising antimalarials against prominent target proteins that have the potential to be a clinical candidate. Here, we also reviewed different aspects of drug resistance and highlighted new drug candidates that are currently in a clinical trial or clinical development, along with a few other molecules with excellent antimalarial activity overs ACTs. The summarized scientific value of previous approaches and structural features of antimalarials related to the activity are highlighted that will be helpful for the development of next-generation antimalarials.
Collapse
Affiliation(s)
- Hari Madhav
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia (A Central University), New Delhi, 110025, India.
| | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia (A Central University), New Delhi, 110025, India.
| |
Collapse
|
8
|
Rylands LI, Welsh A, Maepa K, Stringer T, Taylor D, Chibale K, Smith GS. Structure-activity relationship studies of antiplasmodial cyclometallated ruthenium(II), rhodium(III) and iridium(III) complexes of 2-phenylbenzimidazoles. Eur J Med Chem 2019; 161:11-21. [DOI: 10.1016/j.ejmech.2018.10.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/03/2018] [Accepted: 10/09/2018] [Indexed: 12/24/2022]
|
9
|
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria. Future Med Chem 2018; 10:1853-1874. [PMID: 30019917 DOI: 10.4155/fmc-2017-0250] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Malaria remains one of the most lethal infectious diseases worldwide, and the most severe form is caused by Plasmodium falciparum. In recent decades, the major challenge to treatment of this disease has been the ability of the protozoan parasite to develop resistance to the drugs that are currently in use. Among P. falciparum enzymes, P. falciparum dihydroorotate dehydrogenase has been identified as an important target in drug discovery. Interference with the activity of this enzyme inhibits de novo pyrimidine biosynthesis and consequently prevents malarial infection. Organic synthesis, x-ray crystallography, high-throughput screening and molecular modeling methods such as molecular docking, quantitative structure-activity relationships, structure-based pharmacophore mapping and molecular dynamics simulations have been applied to the discovery of new inhibitors of P. falciparum dihydroorotate dehydrogenase.
Collapse
|
10
|
Moreno-Sabater A, Pérignon JL, Mazier D, Lavazec C, Soulard V. Humanized mouse models infected with human Plasmodium species for antimalarial drug discovery. Expert Opin Drug Discov 2017; 13:131-140. [DOI: 10.1080/17460441.2018.1410136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Alicia Moreno-Sabater
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
- Assistance Publique - Hopitaux de Paris - Hôpitaux Universitaires Paris-Est - Site Saint-Antoine, Paris, Île-de-France France
| | | | - Dominique Mazier
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
| | - Catherine Lavazec
- Institut Cochin – INSERM U1016, Paris, Île-de-France France
- CNRS - UMR8104, Paris, France
- Universite Paris Descartes, Paris, Île-de-France France
| | - Valerie Soulard
- UPMC Faculte de Medecine - INSERM U1135, CNRS ERL 8255, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, Île-de-France France
| |
Collapse
|
11
|
Arnould S, Rodier G, Matar G, Vincent C, Pirot N, Delorme Y, Berthet C, Buscail Y, Noël JY, Lachambre S, Jarlier M, Bernex F, Delpech H, Vidalain PO, Janin YL, Theillet C, Sardet C. Checkpoint kinase 1 inhibition sensitises transformed cells to dihydroorotate dehydrogenase inhibition. Oncotarget 2017; 8:95206-95222. [PMID: 29221122 PMCID: PMC5707016 DOI: 10.18632/oncotarget.19199] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/17/2017] [Indexed: 12/17/2022] Open
Abstract
Reduction in nucleotide pools through the inhibition of mitochondrial enzyme dihydroorotate dehydrogenase (DHODH) has been demonstrated to effectively reduce cancer cell proliferation and tumour growth. The current study sought to investigate whether this antiproliferative effect could be enhanced by combining Chk1 kinase inhibition. The pharmacological activity of DHODH inhibitor teriflunomide was more selective towards transformed mouse embryonic fibroblasts than their primary or immortalised counterparts, and this effect was amplified when cells were subsequently exposed to PF477736 Chk1 inhibitor. Flow cytometry analyses revealed substantial accumulations of cells in S and G2/M phases, followed by increased cytotoxicity which was characterised by caspase 3-dependent induction of cell death. Associating PF477736 with teriflunomide also significantly sensitised SUM159 and HCC1937 human triple negative breast cancer cell lines to dihydroorotate dehydrogenase inhibition. The main characteristic of this effect was the sustained accumulation of teriflunomide-induced DNA damage as cells displayed increased phospho serine 139 H2AX (γH2AX) levels and concentration-dependent phosphorylation of Chk1 on serine 345 upon exposure to the combination as compared with either inhibitor alone. Importantly a similar significant increase in cell death was observed upon dual siRNA mediated depletion of Chk1 and DHODH in both murine and human cancer cell models. Altogether these results suggest that combining DHODH and Chk1 inhibitions may be a strategy worth considering as a potential alternative to conventional chemotherapies.
Collapse
Affiliation(s)
- Stéphanie Arnould
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Geneviève Rodier
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Gisèle Matar
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Charles Vincent
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Nelly Pirot
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
- Réseau d'Histologie Expérimentale de Montpellier, BioCampus, UMS3426 CNRS-US009 INSERM-UM, Montpellier, France
| | - Yoann Delorme
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Charlène Berthet
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
- Réseau d'Histologie Expérimentale de Montpellier, BioCampus, UMS3426 CNRS-US009 INSERM-UM, Montpellier, France
| | - Yoan Buscail
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
- Réseau d'Histologie Expérimentale de Montpellier, BioCampus, UMS3426 CNRS-US009 INSERM-UM, Montpellier, France
| | - Jean Yohan Noël
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
- Réseau d'Histologie Expérimentale de Montpellier, BioCampus, UMS3426 CNRS-US009 INSERM-UM, Montpellier, France
| | - Simon Lachambre
- Montpellier RIO Imaging, BioCampus, UMS3426 CNRS-US009 INSERM-UM, Montpellier, France
| | - Marta Jarlier
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Florence Bernex
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
- Réseau d'Histologie Expérimentale de Montpellier, BioCampus, UMS3426 CNRS-US009 INSERM-UM, Montpellier, France
| | - Hélène Delpech
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Pierre Olivier Vidalain
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Equipe Chimie and Biologie, Modélisation et Immunologie pour la Thérapie, CNRS UMR 8601 CNRS-Université Paris Descartes, Paris, France
| | - Yves L. Janin
- Institut Pasteur, Unité de Chimie et Biocatalyse, CNRS UMR3523, Paris, France
| | - Charles Theillet
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Claude Sardet
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
- INSERM U1194, Montpellier, France
- Université de Montpellier, Montpellier, France
- Institut Régional du Cancer de Montpellier, Montpellier, France
| |
Collapse
|
12
|
+Targeting Mitochondrial Functions as Antimalarial Regime, What Is Next? CURRENT CLINICAL MICROBIOLOGY REPORTS 2017. [DOI: 10.1007/s40588-017-0075-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
2-Aminothiophene scaffolds: Diverse biological and pharmacological attributes in medicinal chemistry. Eur J Med Chem 2017; 140:465-493. [PMID: 28987607 DOI: 10.1016/j.ejmech.2017.09.039] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/02/2017] [Accepted: 09/19/2017] [Indexed: 12/30/2022]
Abstract
2-Aminothiophenes are important five-membered heterocyclic building blocks in organic synthesis, and the chemistry of these small molecules is still developing based on the discovery of cyclization by Gewald. Another attractive feature of 2-aminothiophene scaffolds is their ability to act as synthons for the synthesis of biological active thiophene-containing heterocycles, conjugates and hybrids. Currently, the biological actions of 2-aminothiophenes or their 2-N-substituted analogues are still being investigated because of their various mechanisms of action (e.g., pharmacophore and pharmacokinetic properties). Likewise, the 2-aminothiophene family is used as diverse promising selective inhibitors, receptors, and modulators in medicinal chemistry, and these compounds even exhibit effective pharmacological properties in the various clinical phases of appropriate diseases. In this review, major biological and pharmacological reports on 2-aminothiophenes and related compounds have been highlighted; most perspective drug-candidate hits were selected for discussion and described, along with additional synthetic pathways. In addition, we focused on the literature dedicated to 2-aminothiophenes and 2-N-substituted derivatives, which have been published from 2010 to 2017.
Collapse
|
14
|
Bu FZ, Tan XJ, Xing DX, Wang C. Design, synthesis, crystal structure and in vitro cytotoxic properties of a novel Schiff base derived from indole and biphenyl. ACTA CRYSTALLOGRAPHICA SECTION C-STRUCTURAL CHEMISTRY 2017; 73:546-555. [DOI: 10.1107/s2053229617009044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/16/2017] [Indexed: 03/18/2023]
Abstract
A novel and potentially active dihydroorotate dehydrogenase (DHODH) inhibitor, namely 3-({(E)-[(E)-1-(biphenyl-4-yl)ethylidene]hydrazinylidene}methyl)-1H-indole (BEHI) acetonitrile disolvate, C23H19N3·2CH3CN, has been designed and synthesized. The structure of BEHI was characterized by elemental analysis, Q-TOF (quadrupole time-of-flight) MS, NMR, UV–Vis and single-crystal X-ray diffraction. The antitumour activity of the target molecule was evaluated by the MTT method. Results indicated that BEHI exhibited rather potent cytotoxic activity against human A549 (IC50 = 20.5 µM) and mouse breast 4T1 (IC50 = 18.5 µM) cancer cell lines. Meanwhile, to rationalize its potencies in the target, BEHI was docked into DHODH and the interactions with the active site residues were analyzed. Single-crystal structure analysis indicated that hydrogen bonds are present only between BEHI and acetonitrile solvent molecules in the asymmetric unit. The interplay of weak π–π stacking and weak C(N)—H...π interactions between neighbouring BEHI molecules play crucial roles in the formation of the final supramolecular frameworks.
Collapse
|
15
|
Vargas-Oviedo D, Charris-Molina A, Portilla J. Efficient Access to o
-Phenylendiamines and Their Use in the Synthesis of a 1,2-Dialkyl-5-trifluoromethylbenzimidazoles Library Under Microwave Conditions. ChemistrySelect 2017. [DOI: 10.1002/slct.201700623] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Diana Vargas-Oviedo
- Departamento de Química; Universidad de los Andes; Carrera 1 N° 18A-12 Bogotá Colombia
| | - Andrés Charris-Molina
- Departamento de Química; Universidad de los Andes; Carrera 1 N° 18A-12 Bogotá Colombia
| | - Jaime Portilla
- Departamento de Química; Universidad de los Andes; Carrera 1 N° 18A-12 Bogotá Colombia
| |
Collapse
|
16
|
Singh A, Maqbool M, Mobashir M, Hoda N. Dihydroorotate dehydrogenase: A drug target for the development of antimalarials. Eur J Med Chem 2017; 125:640-651. [DOI: 10.1016/j.ejmech.2016.09.085] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/07/2016] [Accepted: 09/25/2016] [Indexed: 02/03/2023]
|
17
|
Vyas VK, Qureshi G, Ghate M, Patel H, Dalai S. Identification of novel PfDHODH inhibitors as antimalarial agents via pharmacophore-based virtual screening followed by molecular docking and in vivo antimalarial activity. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2016; 27:427-440. [PMID: 27310104 DOI: 10.1080/1062936x.2016.1189959] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/11/2016] [Indexed: 06/06/2023]
Abstract
Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) catalyses the fourth reaction of de novo pyrimidine biosynthesis in parasites, and represents an important target for the treatment of malaria. In this study, we describe pharmacophore-based virtual screening combined with docking study and biological evaluation as a rational strategy for identification of novel hits as antimalarial agents. Pharmacophore models were established from known PfDHODH inhibitors using the GALAHAD module with IC50 values ranging from 0.033 μM to 142 μM. The best pharmacophore model consisted of three hydrogen bond acceptor, one hydrogen bond donor and one hydrophobic features. The pharmacophore models were validated through receiver operating characteristic and Günere-Henry scoring methods. The best pharmacophore model as a 3D search query was searched against the IBS database. Several compounds with different structures (scaffolds) were retrieved as hit molecules. Among these compounds, those with a QFIT value of more than 81 were docked in the PfDHODH enzyme to further explore the binding modes of these compounds. In silico pharmacokinetic and toxicities were predicted for the best docked molecules. Finally, the identified hits were evaluated in vivo for their antimalarial activity in a parasite inhibition assay. The hits reported here showed good potential to become novel antimalarial agents.
Collapse
Affiliation(s)
- V K Vyas
- a Department of Pharmaceutical Chemistry , Institute of Pharmacy, Nirma University , Ahmedabad , India
| | - G Qureshi
- a Department of Pharmaceutical Chemistry , Institute of Pharmacy, Nirma University , Ahmedabad , India
| | - M Ghate
- a Department of Pharmaceutical Chemistry , Institute of Pharmacy, Nirma University , Ahmedabad , India
| | - H Patel
- b Institute of Science, Nirma University , Ahmedabad , India
| | - S Dalai
- b Institute of Science, Nirma University , Ahmedabad , India
| |
Collapse
|
18
|
Hou X, Chen X, Zhang M, Yan A. QSAR study on the antimalarial activity of Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) inhibitors. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2016; 27:101-124. [PMID: 26911561 DOI: 10.1080/1062936x.2015.1134652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Plasmodium falciparum, the most fatal parasite that causes malaria, is responsible for over one million deaths per year. P. falciparum dihydroorotate dehydrogenase (PfDHODH) has been validated as a promising drug development target for antimalarial therapy since it catalyzes the rate-limiting step for DNA and RNA biosynthesis. In this study, we investigated the quantitative structure-activity relationships (QSAR) of the antimalarial activity of PfDHODH inhibitors by generating four computational models using a multilinear regression (MLR) and a support vector machine (SVM) based on a dataset of 255 PfDHODH inhibitors. All the models display good prediction quality with a leave-one-out q(2) >0.66, a correlation coefficient (r) >0.85 on both training sets and test sets, and a mean square error (MSE) <0.32 on training sets and <0.37 on test sets, respectively. The study indicated that the hydrogen bonding ability, atom polarizabilities and ring complexity are predominant factors for inhibitors' antimalarial activity. The models are capable of predicting inhibitors' antimalarial activity and the molecular descriptors for building the models could be helpful in the development of new antimalarial drugs.
Collapse
Affiliation(s)
- X Hou
- a State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering , Beijing University of Chemical Technology , Beijing , P.R. China
| | - X Chen
- a State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering , Beijing University of Chemical Technology , Beijing , P.R. China
| | - M Zhang
- a State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering , Beijing University of Chemical Technology , Beijing , P.R. China
| | - A Yan
- a State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering , Beijing University of Chemical Technology , Beijing , P.R. China
- b Stake Key Laboratory of Natural and Biomimetic Drugs , Peking University , Beijing , P.R. China
| |
Collapse
|
19
|
Aneja B, Kumar B, Jairajpuri MA, Abid M. A structure guided drug-discovery approach towards identification of Plasmodium inhibitors. RSC Adv 2016. [DOI: 10.1039/c5ra19673f] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
This article provides a comprehensive review of inhibitors from natural, semisynthetic or synthetic sources against key targets ofPlasmodium falciparum.
Collapse
Affiliation(s)
- Babita Aneja
- Medicinal Chemistry Lab
- Department of Biosciences
- Jamia Millia Islamia (A Central University)
- New Delhi 110025
- India
| | - Bhumika Kumar
- Medicinal Chemistry Lab
- Department of Biosciences
- Jamia Millia Islamia (A Central University)
- New Delhi 110025
- India
| | - Mohamad Aman Jairajpuri
- Protein Conformation and Enzymology Lab
- Department of Biosciences
- Jamia Millia Islamia (A Central University)
- New Delhi 110025
- India
| | - Mohammad Abid
- Medicinal Chemistry Lab
- Department of Biosciences
- Jamia Millia Islamia (A Central University)
- New Delhi 110025
- India
| |
Collapse
|
20
|
Copper-mediated N-Arylation of Methyl 2-Aminothiophene-3-carboxylate with Organoboron Reagents. Tetrahedron Lett 2015; 56:6839-6842. [PMID: 26576065 DOI: 10.1016/j.tetlet.2015.10.080] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
A practical protocol for the synthesis of N-arylated methyl 2-aminothiophene-3-carboxylate has been developed via Chan-Lam cross-coupling. The desired products were synthesized by cross-coupling of methyl 2-aminothiophene-3-carboxylate with both arylboronic acids and potassium aryltrifluoroborate salts in moderate to good yields. A broad range of functional groups was well tolerated.
Collapse
|
21
|
Lucas-Hourani M, Munier-Lehmann H, El Mazouni F, Malmquist NA, Harpon J, Coutant EP, Guillou S, Helynck O, Noel A, Scherf A, Phillips MA, Tangy F, Vidalain PO, Janin YL. Original 2-(3-Alkoxy-1H-pyrazol-1-yl)azines Inhibitors of Human Dihydroorotate Dehydrogenase (DHODH). J Med Chem 2015; 58:5579-98. [PMID: 26079043 PMCID: PMC4516315 DOI: 10.1021/acs.jmedchem.5b00606] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Following our discovery of human dihydroorotate dehydrogenase (DHODH) inhibition by 2-(3-alkoxy-1H-pyrazol-1-yl)pyrimidine derivatives as well as 2-(4-benzyl-3-ethoxy-5-methyl-1H-pyrazol-1-yl)-5-methylpyridine, we describe here the syntheses and evaluation of an array of azine-bearing analogues. As in our previous report, the structure-activity study of this series of human DHODH inhibitors was based on a phenotypic assay measuring measles virus replication. Among other inhibitors, this round of syntheses and biological evaluation iteration led to the highly active 5-cyclopropyl-2-(4-(2,6-difluorophenoxy)-3-isopropoxy-5-methyl-1H-pyrazol-1-yl)-3-fluoropyridine. Inhibition of DHODH by this compound was confirmed in an array of in vitro assays, including enzymatic tests and cell-based assays for viral replication and cellular growth. This molecule was found to be more active than the known inhibitors of DHODH, brequinar and teriflunomide, thus opening perspectives for its use as a tool or for the design of an original series of immunosuppressive agent. Moreover, because other series of inhibitors of human DHODH have been found to also affect Plasmodium falciparum DHODH, all the compounds were assayed for their effect on P. falciparum growth. However, the modest in vitro inhibition solely observed for two compounds did not correlate with their inhibition of P. falciparum DHODH.
Collapse
Affiliation(s)
- Marianne Lucas-Hourani
- †Unité de Génomique Virale et Vaccination, Département de Virologie, Institut Pasteur, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,‡Unité Mixte de Recherche 3569, Centre National de la Recherche Scientifique, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Hélène Munier-Lehmann
- §Unité Mixte de Recherche 3523, Centre National de la Recherche Scientifique, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,∥Unité de Chimie et Biocatalyse, Département de Biologie Structurale et Chimie, Institut Pasteur, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Farah El Mazouni
- ⊥Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 6001 Forest Park Boulevard, Dallas, Texas 75390-9041, United States
| | - Nicholas A Malmquist
- #Unité de Biologie des Interactions Hôte-Parasite, Département de Parasitologie et Mycologie, Institut Pasteur, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France.,^Unité 1201, Institut National de la Santé et de la Recherche Médicale, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,+Equipe de Recherche Labellisée 9195, Centre National de la Recherche Scientifique, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Jane Harpon
- #Unité de Biologie des Interactions Hôte-Parasite, Département de Parasitologie et Mycologie, Institut Pasteur, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France.,^Unité 1201, Institut National de la Santé et de la Recherche Médicale, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,+Equipe de Recherche Labellisée 9195, Centre National de la Recherche Scientifique, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Eloi P Coutant
- §Unité Mixte de Recherche 3523, Centre National de la Recherche Scientifique, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,∥Unité de Chimie et Biocatalyse, Département de Biologie Structurale et Chimie, Institut Pasteur, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Sandrine Guillou
- §Unité Mixte de Recherche 3523, Centre National de la Recherche Scientifique, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,∥Unité de Chimie et Biocatalyse, Département de Biologie Structurale et Chimie, Institut Pasteur, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Olivier Helynck
- §Unité Mixte de Recherche 3523, Centre National de la Recherche Scientifique, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,∥Unité de Chimie et Biocatalyse, Département de Biologie Structurale et Chimie, Institut Pasteur, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Anne Noel
- §Unité Mixte de Recherche 3523, Centre National de la Recherche Scientifique, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,∥Unité de Chimie et Biocatalyse, Département de Biologie Structurale et Chimie, Institut Pasteur, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Artur Scherf
- #Unité de Biologie des Interactions Hôte-Parasite, Département de Parasitologie et Mycologie, Institut Pasteur, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France.,^Unité 1201, Institut National de la Santé et de la Recherche Médicale, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,+Equipe de Recherche Labellisée 9195, Centre National de la Recherche Scientifique, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Margaret A Phillips
- ⊥Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, 6001 Forest Park Boulevard, Dallas, Texas 75390-9041, United States
| | - Frédéric Tangy
- †Unité de Génomique Virale et Vaccination, Département de Virologie, Institut Pasteur, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,‡Unité Mixte de Recherche 3569, Centre National de la Recherche Scientifique, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Pierre-Olivier Vidalain
- †Unité de Génomique Virale et Vaccination, Département de Virologie, Institut Pasteur, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,‡Unité Mixte de Recherche 3569, Centre National de la Recherche Scientifique, 25 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Yves L Janin
- §Unité Mixte de Recherche 3523, Centre National de la Recherche Scientifique, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France.,∥Unité de Chimie et Biocatalyse, Département de Biologie Structurale et Chimie, Institut Pasteur, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| |
Collapse
|
22
|
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: 206] [Impact Index Per Article: 20.6] [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
|
23
|
Targeting the mitochondrial electron transport chain of Plasmodium falciparum: new strategies towards the development of improved antimalarials for the elimination era. Future Med Chem 2014; 5:1573-91. [PMID: 24024949 DOI: 10.4155/fmc.13.121] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Despite intense efforts, there has not been a truly new antimalarial, possessing a novel mechanism of action, registered for over 10 years. By virtue of a novel mode of action, it is hoped that the global challenge of multidrug-resistant parasites can be overcome, as well as developing drugs that possess prophylaxis and/or transmission-blocking properties, towards an elimination agenda. Many target-based and whole-cell screening drug development programs have been undertaken in recent years and here an overview of specific projects that have focused on targeting the parasite's mitochondrial electron transport chain is presented. Medicinal chemistry activity has largely focused on inhibitors of the parasite cytochrome bc1 Complex (Complex III) including acridinediones, pyridones and quinolone aryl esters, as well as inhibitors of dihydroorotate dehydrogenase that includes triazolopyrimidines and benzimidazoles. Common barriers to progress and opportunities for novel chemistry and potential additional electron transport chain targets are discussed in the context of the target candidate profiles for uncomplicated malaria.
Collapse
|
24
|
Harnessing evolutionary fitness in Plasmodium falciparum for drug discovery and suppressing resistance. Proc Natl Acad Sci U S A 2013; 111:799-804. [PMID: 24381157 DOI: 10.1073/pnas.1320886110] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Drug resistance emerges in an ecological context where fitness costs restrict the diversity of escape pathways. These pathways are targets for drug discovery, and here we demonstrate that we can identify small-molecule inhibitors that differentially target resistant parasites. Combining wild-type and mutant-type inhibitors may prevent the emergence of competitively viable resistance. We tested this hypothesis with a clinically derived chloroquine-resistant (CQ(r)) malaria parasite and with parasites derived by in vitro selection with Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) inhibitors. We screened a chemical library against CQ(s) and CQ(r) lines and discovered a drug-like compound (IDI-3783) that was potent only in the CQ(r) line. Surprisingly, in vitro selection of Plasmodium falciparum resistant to IDI-3783 restored CQ sensitivity, thereby indicating that CQ might once again be useful as a malaria therapy. In parallel experiments, we selected P. falciparum lines resistant to structurally unrelated PfDHODH inhibitors (Genz-666136 and DSM74). Both selections yielded resistant lines with the same point mutation in PfDHODH:E182D. We discovered a compound (IDI-6273) more potent against E182D than wild-type parasites. Selection of the E182D mutant with IDI-6273 yielded a reversion to the wild-type protein sequence and phenotype although the nucleotide sequence was different. Importantly, selection with a combination of Genz-669178, a wild-type PfDHODH inhibitor, and IDI-6273, a mutant-selective PfDHODH inhibitor, did not yield resistant parasites. These two examples demonstrate that the compromise between resistance and evolutionary fitness can be exploited to design therapies that prevent the emergence and spread of resistant organisms.
Collapse
|
25
|
Banning JE, Gentillon J, Ryabchuk PG, Prosser AR, Rogers A, Edwards A, Holtzen A, Babkov IA, Rubina M, Rubin M. Formal Substitution of Bromocyclopropanes with Nitrogen Nucleophiles. J Org Chem 2013; 78:7601-16. [DOI: 10.1021/jo4011798] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Joseph E. Banning
- Department of Chemistry, The University
of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045-75832
| | - Jacob Gentillon
- Department of Chemistry, The University
of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045-75832
| | - Pavel G. Ryabchuk
- Department of Chemistry, The University
of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045-75832
| | - Anthony R. Prosser
- Department of Chemistry, The University
of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045-75832
| | - Andrew Rogers
- Department of Chemistry, The University
of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045-75832
| | - Andrew Edwards
- Department of Chemistry, The University
of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045-75832
| | - Andrew Holtzen
- Department of Chemistry, The University
of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045-75832
| | - Ivan A. Babkov
- Department of Chemistry, The University
of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045-75832
| | - Marina Rubina
- Department of Chemistry, The University
of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045-75832
| | - Michael Rubin
- Department of Chemistry, The University
of Kansas, 1251 Wescoe Hall Drive, Lawrence, Kansas 66045-75832
| |
Collapse
|
26
|
Abstract
The emergence of resistance to artemisinins and the renewed efforts to eradicate malaria demand the urgent development of new drugs. In this endeavour, the evaluation of efficacy in animal models is often a go/no go decision assay in drug discovery. This important role relies on the capability of animal models to assess the disposition, toxicology and efficacy of drugs in a single test. Although the relative merits of each efficacy model of malaria as human surrogate have been extensively discussed, there are no critical analyses on the use of such models in current drug discovery. In this article, we intend to analyse how efficacy models are used to discover new antimalarial drugs. Our analysis indicates that testing drug efficacy is often the last assay in each discovery stage and the experimental designs utilized are not optimized to expedite decision-making and inform clinical development. In light of this analysis, we propose new ways to accelerate drug discovery using efficacy models.
Collapse
|
27
|
Biamonte MA, Wanner J, Le Roch KG. Recent advances in malaria drug discovery. Bioorg Med Chem Lett 2013; 23:2829-43. [PMID: 23587422 PMCID: PMC3762334 DOI: 10.1016/j.bmcl.2013.03.067] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/11/2013] [Accepted: 03/20/2013] [Indexed: 01/18/2023]
Abstract
This digest covers some of the most relevant progress in malaria drug discovery published between 2010 and 2012. There is an urgent need to develop new antimalarial drugs. Such drugs can target the blood stage of the disease to alleviate the symptoms, the liver stage to prevent relapses, and the transmission stage to protect other humans. The pipeline for the blood stage is becoming robust, but this should not be a source of complacency, as the current therapies set a high standard. Drug discovery efforts directed towards the liver and transmission stages are in their infancy but are receiving increasing attention as targeting these stages could be instrumental in eradicating malaria.
Collapse
Affiliation(s)
- Marco A Biamonte
- Drug Discovery for Tropical Diseases, Suite 230, San Diego, CA 92121, USA.
| | | | | |
Collapse
|
28
|
Munier-Lehmann H, Vidalain PO, Tangy F, Janin YL. On dihydroorotate dehydrogenases and their inhibitors and uses. J Med Chem 2013; 56:3148-67. [PMID: 23452331 DOI: 10.1021/jm301848w] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Proper nucleosides availability is crucial for the proliferation of living entities (eukaryotic cells, parasites, bacteria, and virus). Accordingly, the uses of inhibitors of the de novo nucleosides biosynthetic pathways have been investigated in the past. In the following we have focused on dihydroorotate dehydrogenase (DHODH), the fourth enzyme in the de novo pyrimidine nucleosides biosynthetic pathway. We first described the different types of enzyme in terms of sequence, structure, and biochemistry, including the reported bioassays. In a second part, the series of inhibitors of this enzyme along with a description of their potential or actual uses were reviewed. These inhibitors are indeed used in medicine to treat autoimmune diseases such as rheumatoid arthritis or multiple sclerosis (leflunomide and teriflunomide) and have been investigated in treatments of cancer, virus, and parasite infections (i.e., malaria) as well as in crop science.
Collapse
Affiliation(s)
- Hélène Munier-Lehmann
- Institut Pasteur, Unité de Chimie et Biocatalyse, Département de Biologie Structurale et Chimie, 28 Rue du Dr. Roux, 75724 Paris Cedex 15, France
| | | | | | | |
Collapse
|
29
|
Miller MS, Pinson JA, Zheng Z, Jennings IG, Thompson PE. Regioselective synthesis of 5- and 6-methoxybenzimidazole-1,3,5-triazines as inhibitors of phosphoinositide 3-kinase. Bioorg Med Chem Lett 2013; 23:802-5. [DOI: 10.1016/j.bmcl.2012.11.076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 11/19/2012] [Accepted: 11/20/2012] [Indexed: 10/27/2022]
|
30
|
Calderón F, Wilson DM, Gamo FJ. Antimalarial drug discovery: recent progress and future directions. PROGRESS IN MEDICINAL CHEMISTRY 2013; 52:97-151. [PMID: 23384667 DOI: 10.1016/b978-0-444-62652-3.00003-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Affiliation(s)
- Félix Calderón
- Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Spain
| | | | | |
Collapse
|
31
|
Anthony MP, Burrows JN, Duparc S, JMoehrle J, Wells TNC. The global pipeline of new medicines for the control and elimination of malaria. Malar J 2012; 11:316. [PMID: 22958514 PMCID: PMC3472257 DOI: 10.1186/1475-2875-11-316] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/21/2012] [Indexed: 12/03/2022] Open
Abstract
Over the past decade, there has been a transformation in the portfolio of medicines to combat malaria. New fixed-dose artemisinin combination therapy is available, with four different types having received approval from Stringent Regulatory Authorities or the World Health Organization (WHO). However, there is still scope for improvement. The Malaria Eradication Research agenda identified several gaps in the current portfolio. Simpler regimens, such as a single-dose cure are needed, compared with the current three-day treatment. In addition, new medicines that prevent transmission and also relapse are needed, but with better safety profiles than current medicines. There is also a big opportunity for new medicines to prevent reinfection and to provide chemoprotection. This study reviews the global portfolio of new medicines in development against malaria, as of the summer of 2012. Cell-based phenotypic screening, and 'fast followers' of clinically validated classes, mean that there are now many new classes of molecules starting in clinical development, especially for the blood stages of malaria. There remain significant gaps for medicines blocking transmission, preventing relapse, and long-duration molecules for chemoprotection. The nascent pipeline of new medicines is significantly stronger than five years ago. However, there are still risks ahead in clinical development and sustainable funding of clinical studies is vital if this early promise is going to be delivered.
Collapse
Affiliation(s)
- Melinda P Anthony
- Medicines for Malaria Venture (MMV), 20 rte de Pré-Bois 1215, Geneva, Switzerland
| | - Jeremy N Burrows
- Medicines for Malaria Venture (MMV), 20 rte de Pré-Bois 1215, Geneva, Switzerland
| | - Stephan Duparc
- Medicines for Malaria Venture (MMV), 20 rte de Pré-Bois 1215, Geneva, Switzerland
| | - Joerg JMoehrle
- Medicines for Malaria Venture (MMV), 20 rte de Pré-Bois 1215, Geneva, Switzerland
| | - Timothy NC Wells
- Medicines for Malaria Venture (MMV), 20 rte de Pré-Bois 1215, Geneva, Switzerland
| |
Collapse
|
32
|
Vyas VK, Parikh H, Ghate M. 3D QSAR studies on 5-(2-methylbenzimidazol-1-yl)-N-alkylthiophene-2-carboxamide derivatives as P. falciparum dihydroorotate dehydrogenase (PfDHODH) inhibitors. Med Chem Res 2012. [DOI: 10.1007/s00044-012-0216-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
33
|
Ryabchuk P, Rubina M, Xu J, Rubin M. Formal Nucleophilic Substitution of Bromocyclopropanes with Azoles. Org Lett 2012; 14:1752-5. [DOI: 10.1021/ol300352z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Pavel Ryabchuk
- Department of Chemistry, University of Kansas 1251 Wescoe Hall Drive, Lawrence, Kansas 66045, United States
| | - Marina Rubina
- Department of Chemistry, University of Kansas 1251 Wescoe Hall Drive, Lawrence, Kansas 66045, United States
| | - Jack Xu
- Department of Chemistry, University of Kansas 1251 Wescoe Hall Drive, Lawrence, Kansas 66045, United States
| | - Michael Rubin
- Department of Chemistry, University of Kansas 1251 Wescoe Hall Drive, Lawrence, Kansas 66045, United States
| |
Collapse
|
34
|
|