1
|
Lima LEBD, Almeida MLGD, Gomes GS, do Nascimento PH, Silva CJOE, da Silva CRD, Tanaka YMR, Romão TP, de Lima TBS, de Araújo ES, de Araújo PLB, Cabral Filho PE, Holanda VN, de Oliveira RN, Figueiredo RCBQD. A phthalimide-triazole derivative obtained by click chemistry exhibits trypanocidal activity, induces autophagy and ameliorates Trypanosoma cruzi infection. Biomed Pharmacother 2025; 186:117963. [PMID: 40101588 DOI: 10.1016/j.biopha.2025.117963] [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: 01/09/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
Chagas disease (CD), caused by Trypanosoma cruzi, remains a leading cause of cardiomyopathy and heart failure in Latin America. Since the 1970s, benznidazole (BNZ) and nifurtimox (NFX) have been the only chemotherapeutic agents used to treat CD. However, their toxicity and low effectiveness in the chronic phase of the disease, make the development of more efficient chemotherapeutics imperative. Here, we investigated the effects of 1,2,3-triazole hybrids, synthesized via click chemistry, containing either phthalimide (FT1, FT2, FT3, FT4) or naphthoquinone (NT1) moieties on T. cruzi and their cytotoxicity on mammalian cells. NT1 and FT1 were the most effective against intracellular parasite with an IC50 = 31.1 and 189.2 µM, respectively. FT1-FT4 showed low cytotoxicity to mammalian cells (CC50 > 754 µM), while NT1 exhibited moderate toxicity (CC50 ≥ 96.1 µM). FT1 demonstrated the highest selectivity towards trypomastigotes and amastigotes with selectivity indexes (SeI) of 6.9 and 6.7, respectively. Ultrastructural analysis of trypomastigotes treated with FT1 revealed mitochondrial alterations, lipid accumulation and Golgi complex disorganization. FT1 also decreased the mitochondrial membrane potential, increased mitochondrial reactive oxygen species (ROS) production, and induced late apoptosis in trypomastigotes. In infected cardiac cells, FT1 treatment led to degradation of amastigotes and Golgi disruption. An increase in autophagosomes in treated host cells and their interaction with intracellular parasites suggest that FT1-induced host cell autophagy may play a role in mitigating the infection and protecting cardiac cells from its deleterious effects.
Collapse
Affiliation(s)
- Lucas Eduardo Bezerra de Lima
- Departamento de Microbiologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Maria Letícia Gomes de Almeida
- Departamento de Microbiologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Gleicyane Silva Gomes
- Departamento de Microbiologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Pedro Henrique do Nascimento
- Departamento de Microbiologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Carla Jasmine Oliveira E Silva
- Departamento de Química, Laboratório de Síntese de Compostos Bioativos, Universidade Federal Rural de Pernambuco, Rua Dom Manoel de Medeiros S/N, Recife, PE 52171900, Brazil
| | - Cecilãne Regina Dioclecia da Silva
- Departamento de Química, Laboratório de Síntese de Compostos Bioativos, Universidade Federal Rural de Pernambuco, Rua Dom Manoel de Medeiros S/N, Recife, PE 52171900, Brazil
| | - Yuri Mouzinho Ramos Tanaka
- Departamento de Entomologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego, s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Tatiany Patrícia Romão
- Departamento de Entomologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego, s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Thaíses Brunelle Santana de Lima
- Departamento de Energia Nuclear, Universidade Federal de Pernambuco (UFPE), Av. Prof. Luiz Freire, 1000, Cidade Universitária, Recife, PE 50740-545, Brazil
| | - Elmo Silvano de Araújo
- Departamento de Energia Nuclear, Universidade Federal de Pernambuco (UFPE), Av. Prof. Luiz Freire, 1000, Cidade Universitária, Recife, PE 50740-545, Brazil
| | - Patricia Lopes Barros de Araújo
- Departamento de Engenharia Biomédica, Universidade Federal de Pernambuco (UFPE), Av. Jornalista Aníbal Fernandes, Cidade Universitária, Recife, PE 50740-560, Brazil
| | - Paulo Euzébio Cabral Filho
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Avenida Prof° Moraes Rego s/n°, Recife, PE 50670901, Brazil
| | - Vanderlan Nogueira Holanda
- Departmento de Biomedicina, Centro Universitário de Vitória de Santo Antão (UNIVISA), Vitória de Santo Antão, PE 55610-050, Brazil
| | - Ronaldo Nascimento de Oliveira
- Departamento de Química, Laboratório de Síntese de Compostos Bioativos, Universidade Federal Rural de Pernambuco, Rua Dom Manoel de Medeiros S/N, Recife, PE 52171900, Brazil.
| | | |
Collapse
|
2
|
Mustière R, Dassonville-Klimpt A, Sonnet P. Aminopyridines in the development of drug candidates against protozoan neglected tropical diseases. Future Med Chem 2024; 16:1357-1373. [PMID: 39109436 PMCID: PMC11318709 DOI: 10.1080/17568919.2024.2359361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/14/2024] [Indexed: 08/15/2024] Open
Abstract
Neglected tropical diseases (NTDs) pose a major threat in tropical zones for impoverished populations. Difficulty of access, adverse effects or low efficacy limit the use of current therapeutic options. Therefore, development of new drugs against NTDs is a necessity. Compounds containing an aminopyridine (AP) moiety are of great interest for the design of new anti-NTD drugs due to their intrinsic properties compared with their closest chemical structures. Currently, over 40 compounds with an AP moiety are on the market, but none is used against NTDs despite active research on APs. The aim of this review is to present the medicinal chemistry work carried out with these scaffolds, against protozoan NTDs: Trypanosoma cruzi, Trypanosoma brucei or Leishmania spp.
Collapse
Affiliation(s)
- Romain Mustière
- Université de Picardie-Jules-Verne, AGIR – Agents infectieux, RéSistance et chimiothérapie, UR 4294, UFR de pharmacie, 1, Rue des Louvels, F-80037 Amiens cedex 1, France
| | - Alexandra Dassonville-Klimpt
- Université de Picardie-Jules-Verne, AGIR – Agents infectieux, RéSistance et chimiothérapie, UR 4294, UFR de pharmacie, 1, Rue des Louvels, F-80037 Amiens cedex 1, France
| | - Pascal Sonnet
- Université de Picardie-Jules-Verne, AGIR – Agents infectieux, RéSistance et chimiothérapie, UR 4294, UFR de pharmacie, 1, Rue des Louvels, F-80037 Amiens cedex 1, France
| |
Collapse
|
3
|
Cristovão-Silva AC, Brelaz-de-Castro MCA, Dionisio da Silva E, Leite ACL, Santiago LBAA, Conceição JMD, da Silva Tiburcio R, de Santana DP, Bedor DCG, de Carvalho BÍV, Ferreira LFGR, de Freitas E Silva R, Alves Pereira VR, Hernandes MZ. Trypanosoma cruzi killing and immune response boosting by novel phenoxyhydrazine-thiazole against Chagas disease. Exp Parasitol 2024; 261:108749. [PMID: 38593864 DOI: 10.1016/j.exppara.2024.108749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/23/2024] [Accepted: 04/06/2024] [Indexed: 04/11/2024]
Abstract
Trypanosoma cruzi (T. cruzi) causes Chagas, which is a neglected tropical disease (NTD). WHO estimates that 6 to 7 million people are infected worldwide. Current treatment is done with benznidazole (BZN), which is very toxic and effective only in the acute phase of the disease. In this work, we designed, synthesized, and characterized thirteen new phenoxyhydrazine-thiazole compounds and applied molecular docking and in vitro methods to investigate cell cytotoxicity, trypanocide activity, nitric oxide (NO) production, cell death, and immunomodulation. We observed a higher predicted affinity of the compounds for the squalene synthase and 14-alpha demethylase enzymes of T. cruzi. Moreover, the compounds displayed a higher predicted affinity for human TLR2 and TLR4, were mildly toxic in vitro for most mammalian cell types tested, and LIZ531 (IC50 2.8 μM) was highly toxic for epimastigotes, LIZ311 (IC50 8.6 μM) for trypomastigotes, and LIZ331 (IC50 1.9 μM) for amastigotes. We observed that LIZ311 (IC50 2.5 μM), LIZ431 (IC50 4.1 μM) and LIZ531 (IC50 5 μM) induced 200 μg/mL of NO and JM14 induced NO production in three different concentrations tested. The compound LIZ331 induced the production of TNF and IL-6. LIZ311 induced the secretion of TNF, IFNγ, IL-2, IL-4, IL-10, and IL-17, cell death by apoptosis, decreased acidic compartment formation, and induced changes in the mitochondrial membrane potential. Taken together, LIZ311 is a promising anti-T. cruzi compound is not toxic to mammalian cells and has increased antiparasitic activity and immunomodulatory properties.
Collapse
Affiliation(s)
- Ana Catarina Cristovão-Silva
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, 50670-42, Recife, Pernambuco, Brazil
| | - Maria Carolina Accioly Brelaz-de-Castro
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, 50670-42, Recife, Pernambuco, Brazil; Laboratory of Parasitology, Vitória Academic Center, Federal University of Pernambuco, 55608-680, Vitória de Santo Antão, Pernambuco, Brazil
| | - Elis Dionisio da Silva
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, 50670-42, Recife, Pernambuco, Brazil
| | - Ana Cristina Lima Leite
- Laboratory of Planning and Synthesis in Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | - Lizandra Beatriz Amorim Alves Santiago
- Laboratory of Planning and Synthesis in Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | - Juliana Maria da Conceição
- Laboratory of Planning and Synthesis in Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | - Robert da Silva Tiburcio
- Laboratory of Planning and Synthesis in Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | - Davi Pereira de Santana
- Pharmaceutical and Cosmetic Development Center (NUDFAC), Department of Pharmaceutical Science, Federal University of Pernambuco, Recife, PE, Brazil
| | - Danilo Cesar Galindo Bedor
- Pharmaceutical and Cosmetic Development Center (NUDFAC), Department of Pharmaceutical Science, Federal University of Pernambuco, Recife, PE, Brazil
| | - Breno Ítalo Valença de Carvalho
- Pharmaceutical and Cosmetic Development Center (NUDFAC), Department of Pharmaceutical Science, Federal University of Pernambuco, Recife, PE, Brazil
| | - Luiz Felipe Gomes Rebello Ferreira
- Laboratory of Theoretical and Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | - Rafael de Freitas E Silva
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, 50670-42, Recife, Pernambuco, Brazil.
| | - Valéria Rêgo Alves Pereira
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, 50670-42, Recife, Pernambuco, Brazil.
| | - Marcelo Zaldini Hernandes
- Laboratory of Theoretical and Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil.
| |
Collapse
|
4
|
Olmo F, Jayawardhana S, Khan AA, Langston HC, Francisco AF, Atherton RL, Ward AI, Taylor MC, Kelly JM, Lewis MD. A panel of phenotypically and genotypically diverse bioluminescent:fluorescent Trypanosoma cruzi strains as a resource for Chagas disease research. PLoS Negl Trop Dis 2024; 18:e0012106. [PMID: 38820564 PMCID: PMC11168640 DOI: 10.1371/journal.pntd.0012106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/12/2024] [Accepted: 05/21/2024] [Indexed: 06/02/2024] Open
Abstract
Chagas disease is caused by Trypanosoma cruzi, a protozoan parasite that displays considerable genetic diversity. Infections result in a range of pathological outcomes, and different strains can exhibit a wide spectrum of anti-parasitic drug tolerance. The genetic determinants of infectivity, virulence and therapeutic susceptibility remain largely unknown. As experimental tools to address these issues, we have generated a panel of bioluminescent:fluorescent parasite strains that cover the diversity of the T. cruzi species. These reporters allow spatio-temporal infection dynamics in murine models to be monitored in a non-invasive manner by in vivo imaging, provide a capability to detect rare infection foci at single-cell resolution, and represent a valuable resource for investigating virulence and host:parasite interactions at a mechanistic level. Importantly, these parasite reporter strains can also contribute to the Chagas disease drug screening cascade by ensuring that candidate compounds have pan-species in vivo activity prior to being advanced into clinical testing. The parasite strains described in this paper are available on request.
Collapse
Affiliation(s)
- Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Archie A. Khan
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Harry C. Langston
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Amanda Fortes Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Richard L. Atherton
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Alex I. Ward
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
5
|
Orlando LMR, Lara LDS, Lechuga GC, Rodrigues GC, Pandoli OG, de Sá DS, Pereira MCDS. Antitrypanosomal Activity of 1,2,3-Triazole-Based Hybrids Evaluated Using In Vitro Preclinical Translational Models. BIOLOGY 2023; 12:1222. [PMID: 37759621 PMCID: PMC10525445 DOI: 10.3390/biology12091222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/08/2023] [Accepted: 07/17/2023] [Indexed: 09/29/2023]
Abstract
Chagas disease therapy still relies on two nitroderivatives, nifurtimox and benznidazole (Bz), which have important limitations and serious adverse effects. New therapeutic alternatives for this silent disease, which has become a worldwide public health problem, are essential for its control and elimination. In this study, 1,2,3-triazole analogues were evaluated for efficacy against T. cruzi. Three triazole derivatives, 1d (0.21 µM), 1f (1.23 µM), and 1g (2.28 µM), showed potent activity against trypomastigotes, reaching IC50 values 10 to 100 times greater than Bz (22.79 µM). Promising candidates are active against intracellular amastigotes (IC50 ≤ 6.20 µM). Treatment of 3D cardiac spheroids, a translational in vitro model, significantly reduced parasite load, indicating good drug diffusion and efficacy. Oral bioavailability was predicted for triazole derivatives. Although infection was significantly reduced without drug pressure in a washout assay, the triazole derivatives did not inhibit parasite resurgence. An isobologram analysis revealed an additive interaction when 1,2,3-triazole analogs and Bz were combined in vitro. These data indicate a strengthened potential of the triazole scaffold and encourage optimization based on an analysis of the structure-activity relationship aimed at identifying new compounds potentially active against T. cruzi.
Collapse
Affiliation(s)
- Lorraine Martins Rocha Orlando
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz Av. Brasil 4365, Rio de Janeiro 21040-900, Brazil; (L.M.R.O.); (L.d.S.L.); (G.C.L.)
| | - Leonardo da Silva Lara
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz Av. Brasil 4365, Rio de Janeiro 21040-900, Brazil; (L.M.R.O.); (L.d.S.L.); (G.C.L.)
| | - Guilherme Curty Lechuga
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz Av. Brasil 4365, Rio de Janeiro 21040-900, Brazil; (L.M.R.O.); (L.d.S.L.); (G.C.L.)
| | - Giseli Capaci Rodrigues
- Programa de Pós-Graduação em Ensino das Ciências, Unigranrio Rua Prof. José de Souza Herdy, Duque de Caxias, Rio de Janeiro 25071-970, Brazil;
| | - Omar Ginoble Pandoli
- Departamento de Química, Pontifícia Universidade Católica, Rua Marquês de São Vincente, 225, Rio de Janeiro 22451-900, Brazil; (O.G.P.); (D.S.d.S.)
- Dipartimento di Farmacia, Università degli Studi di Genova, Viale Cembrano 4, 16126 Genova, Italy
| | - Druval Santos de Sá
- Departamento de Química, Pontifícia Universidade Católica, Rua Marquês de São Vincente, 225, Rio de Janeiro 22451-900, Brazil; (O.G.P.); (D.S.d.S.)
| | - Mirian Claudia de Souza Pereira
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fiocruz Av. Brasil 4365, Rio de Janeiro 21040-900, Brazil; (L.M.R.O.); (L.d.S.L.); (G.C.L.)
| |
Collapse
|
6
|
Wadey GP, Doherty KE, Sandoval AL, Leadbeater NE. Preparation of novel acyl pyrazoles and triazoles by means of oxidative functionalization reactions. HETEROCYCL COMMUN 2023. [DOI: 10.1515/hc-2022-0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
Abstract
Novel acyl pyrazoles and acyl triazoles have been prepared by means of the oxidative amidation of aldehydes in the presence of the requisite azole. Yields range from modest to good in both cases, and some limitations of the substrate scope have been discovered. Acyl pyrazoles were prepared by treatment of a mixture of aldehyde and pyrazole with an oxoammonium salt bearing the nitrate anion. In the case of acyl triazoles, the oxidative functionalization was performed using sodium persulfate as a terminal oxidant in the presence of a catalytic quantity of a nitroxide.
Collapse
Affiliation(s)
- Geoffrey P. Wadey
- Department of Chemistry, University of Connecticut , 55 North Eagleville Road , Storrs , CT 06269 , USA
| | - Katrina E. Doherty
- Department of Chemistry, University of Connecticut , 55 North Eagleville Road , Storrs , CT 06269 , USA
| | - Arturo León Sandoval
- Department of Chemistry, University of Connecticut , 55 North Eagleville Road , Storrs , CT 06269 , USA
| | - Nicholas E. Leadbeater
- Department of Chemistry, University of Connecticut , 55 North Eagleville Road , Storrs , CT 06269 , USA
| |
Collapse
|
7
|
Chen WY, Lin WH, Kuo CJ, Liang CF. Base-mediated ketenimine formation from N-sulfonylthioimidates for the synthesis of 5-amino-1-vinyl/aryl-1,2,3-triazoles. Chem Commun (Camb) 2023; 59:1297-1300. [PMID: 36633138 DOI: 10.1039/d2cc06708k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
N-Sulfonylthioimidate was converted to ketenimine under basic conditions. The reaction with vinyl/aryl azides was induced to cause dipolar cycloaddition to form 5-amino-1-vinyl/aryl-1,2,3-triazoles. The advantages of this method are high efficiency, structural diversity of products favorable yields and applicability to gram-scale operations.
Collapse
Affiliation(s)
- Wan-Yu Chen
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan.
| | - Wei-Han Lin
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan.
| | - Chia-Jou Kuo
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan.
| | - Chien-Fu Liang
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan.
| |
Collapse
|
8
|
Lan J, Cadassou O, Corbet C, Riant O, Feron O. Discovery of Mitochondrial Complex I Inhibitors as Anticancer and Radiosensitizer Drugs Based on Compensatory Stimulation of Lactate Release. Cancers (Basel) 2022; 14:5454. [PMID: 36358872 PMCID: PMC9658316 DOI: 10.3390/cancers14215454] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 08/22/2023] Open
Abstract
Cancer cells may stimulate glycolytic flux when O2 becomes insufficient. Increase in L-lactate release therefore appears as an escape mechanism to drugs targeting mitochondrial respiration but also represents a response that may be exploited to screen for compounds blocking either mitochondrial carriers of oxidizable substrates or the electron transport chain. Here, we developed a screening procedure based on the capacity of cancer cells to release L-lactate to gain insights on the development of mitochondrial complex I inhibitors. For this purpose, we synthesized derivatives of carboxyamidotriazole, a compound previously described as a potential OXPHOS inhibitor. Two series of derivatives were generated by cycloaddition between benzylazide and either cyanoacetamides or alkynes. A primary assay measuring L-lactate release as a compensatory mechanism upon OXPHOS inhibition led us to identify 15 hits among 28 derivatives. A secondary assay measuring O2 consumption in permeabilized cancer cells confirmed that 12 compounds among the hits exhibited reversible complex I inhibitory activity. Anticancer effects of a short list of 5 compounds identified to induce more L-lactate release than reference compound were then evaluated on cancer cells and tumor-mimicking 3D spheroids. Human and mouse cancer cell monolayers exhibiting high level of respiration in basal conditions were up to 3-fold more sensitive than less oxidative cancer cells. 3D tumor spheroids further revealed potency differences between selected compounds in terms of cytotoxicity but also radiosensitizing activity resulting from local reoxygenation. In conclusion, this study documents the feasibility to efficiently screen in 96-well plate format for mitochondrial complex I inhibitors based on the capacity of drug candidates to induce L-lactate release.
Collapse
Affiliation(s)
- Junjie Lan
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
- Institute of Condensed Matter and Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Université catholique de Louvain (UCLouvain), 1348 Louvain-la-Neuve, Belgium
| | - Octavia Cadassou
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Olivier Riant
- Institute of Condensed Matter and Nanosciences (IMCN), Molecular Chemistry, Materials and Catalysis (MOST), Université catholique de Louvain (UCLouvain), 1348 Louvain-la-Neuve, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, avenue Pasteur 6, 1300 Wavre, Belgium
| |
Collapse
|
9
|
Lengerli D, Ibis K, Nural Y, Banoglu E. The 1,2,3-triazole 'all-in-one' ring system in drug discovery: a good bioisostere, a good pharmacophore, a good linker, and a versatile synthetic tool. Expert Opin Drug Discov 2022; 17:1209-1236. [PMID: 36164263 DOI: 10.1080/17460441.2022.2129613] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The 1,2,3-triazole ring occupies an important space in medicinal chemistry due to its unique structural properties, synthetic versatility and pharmacological potential making it a critical scaffold. Since it is readily available through click chemistry for creating compound collections against various diseases, it has become an emerging area of interest for medicinal chemists. AREAS COVERED This review article addresses the unique properties of the1,2,3-triazole nucleus as an intriguing ring system in drug discovery while focusing on the most recent medicinal chemistry strategies exploited for the design and development of 1,2,3-triazole analogs as inhibitors of various biological targets. EXPERT OPINION Evidently, the 1,2,3-triazole ring with unique structural features has enormous potential in drug design against various diseases as a pharmacophore, a bioisoster or a structural platform. The most recent evidence indicates that it may be more emerging in drug molecules in near future along with an increasing understanding of its prominent roles in drug structures. The synthetic feasibility and versatility of triazole chemistry make it certainly ideal for creating compound libraries for more constructive structure-activity relationship studies. However, more comparative and target-specific studies are needed to gain a deeper understanding of the roles of the 1,2,3-triazole ring in molecular recognition.[Figure: see text].
Collapse
Affiliation(s)
- Deniz Lengerli
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Kübra Ibis
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Yahya Nural
- Department of Analytical Chemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Erden Banoglu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| |
Collapse
|
10
|
Temporal and Wash-Out Studies Identify Medicines for Malaria Venture Pathogen Box Compounds with Fast-Acting Activity against Both Trypanosoma cruzi and Trypanosoma brucei. Microorganisms 2022; 10:microorganisms10071287. [PMID: 35889006 PMCID: PMC9317670 DOI: 10.3390/microorganisms10071287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Chagas disease caused by the protozoan Trypanosoma cruzi is endemic to 21 countries in the Americas, effects approximately 6 million people and on average results in 12,000 deaths annually. Human African Trypanosomiasis (HAT) is caused by the Trypanosoma brucei sub-species, endemic to 36 countries within sub-Saharan Africa. Treatment regimens for these parasitic diseases are complicated and not effective against all disease stages; thus, there is a need to find improved treatments. To identify new molecules for the drug discovery pipelines for these diseases, we have utilised in vitro assays to identify compounds with selective activity against both T. cruzi and T.b. brucei from the Medicines for Malaria Venture (MMV) Pathogen Box compound collection. To prioritise these molecules for further investigation, temporal and wash off assays were utilised to identify the speed of action and cidality of compounds. For translational relevance, compounds were tested against clinically relevant T.b. brucei subspecies. Compounds with activity against T. cruzi cytochrome P450 (TcCYP51) have not previously been successful in clinical trials for chronic Chagas disease; thus, to deprioritise compounds with this activity, they were tested against recombinant TcCYP51. Compounds with biological profiles warranting progression offer important tools for drug and target development against kinetoplastids.
Collapse
|
11
|
Mathias F, Kabri Y, Brun D, Primas N, Di Giorgio C, Vanelle P. Synthesis and Anti- Trypanosoma cruzi Biological Evaluation of Novel 2-Nitropyrrole Derivatives. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072163. [PMID: 35408570 PMCID: PMC9000427 DOI: 10.3390/molecules27072163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/17/2022] [Accepted: 03/25/2022] [Indexed: 01/25/2023]
Abstract
Human American trypanosomiasis, called Chagas disease, caused by T. cruzi protozoan infection, represents a major public health problem, with about 7000 annual deaths in Latin America. As part of the search for new and safe anti-Trypanosoma cruzi derivatives involving nitroheterocycles, we report herein the synthesis of ten 1-substituted 2-nitropyrrole compounds and their biological evaluation. After an optimization phase, a convergent synthesis methodology was used to obtain these new final compounds in two steps from the 2-nitropyrrole starting product. All the designed derivatives follow Lipinski’s rule of five. The cytotoxicity evaluation on CHO cells showed no significant cytotoxicity, except for compound 3 (CC50 = 24.3 µM). Compound 18 appeared to show activity against T. cruzi intracellular amastigotes form (EC50 = 3.6 ± 1.8 µM) and good selectivity over the vero host cells. Unfortunately, this compound 18 showed an insufficient maximum effect compared to the reference drug (nifurtimox). Whether longer duration treatments may eliminate all parasites remains to be explored.
Collapse
Affiliation(s)
- Fanny Mathias
- Equipe Pharmaco-Chimie Radicalaire, CNRS, ICR UMR 7273, Faculté de Pharmacie, Aix Marseille University, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (F.M.); (Y.K.); (D.B.); (N.P.)
- Assistance Publique-Hôpitaux de Marseille (APHM), Pharmacie Usage Intérieur, Hôpital Nord, Chemin-des-Bourrely, 13015 Marseille, France
| | - Youssef Kabri
- Equipe Pharmaco-Chimie Radicalaire, CNRS, ICR UMR 7273, Faculté de Pharmacie, Aix Marseille University, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (F.M.); (Y.K.); (D.B.); (N.P.)
| | - Damien Brun
- Equipe Pharmaco-Chimie Radicalaire, CNRS, ICR UMR 7273, Faculté de Pharmacie, Aix Marseille University, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (F.M.); (Y.K.); (D.B.); (N.P.)
| | - Nicolas Primas
- Equipe Pharmaco-Chimie Radicalaire, CNRS, ICR UMR 7273, Faculté de Pharmacie, Aix Marseille University, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (F.M.); (Y.K.); (D.B.); (N.P.)
- Assistance Publique-Hôpitaux de Marseille (APHM), Service Central de la Qualité et de l’Information Pharmaceutiques (SCQIP), Hôpital de la Conception, 147, Boulevard Baille, 13005 Marseille, France
| | - Carole Di Giorgio
- Laboratoire de Mutagénèse Environnementale, CNRS, IRD, Aix Marseille University, IMBE UMR 7263, Avignon University, 13385 Marseille, France;
| | - Patrice Vanelle
- Equipe Pharmaco-Chimie Radicalaire, CNRS, ICR UMR 7273, Faculté de Pharmacie, Aix Marseille University, 27 Boulevard Jean Moulin, CS30064, CEDEX 05, 13385 Marseille, France; (F.M.); (Y.K.); (D.B.); (N.P.)
- Assistance Publique-Hôpitaux de Marseille (APHM), Service Central de la Qualité et de l’Information Pharmaceutiques (SCQIP), Hôpital de la Conception, 147, Boulevard Baille, 13005 Marseille, France
- Correspondence: ; Tel.: +33-4-9183-5580
| |
Collapse
|
12
|
Franco CA, da Silva TI, Dias MG, Ferreira BW, de Sousa BL, Bousada GM, Barreto RW, Vaz BG, Lima GDS, Dos Santos MH, Grossi JAS, Vieira Varejão EV. Synthesis of Tyrosol 1,2,3-Triazole Derivatives and Their Phytotoxic Activity against Euphorbia heterophylla. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:2806-2816. [PMID: 35225607 DOI: 10.1021/acs.jafc.1c06012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The synthesis and phytotoxic activity of a series of tyrosol 1,2,3-triazole derivatives are reported herein. Target compounds were synthesized through the copper(I)-catalyzed azide-alkyne cycloaddition reaction (CuAAC), known as click reaction, and these were tested for phytotoxic activity on leaves of wild poinsettia (Euphorbia heterophylla), fleabane (Conyza sumatrensis), and tropical spiderwort (Commelina benghalensis). These are three highly noxious agricultural weeds that challenge available weed control methods, including the use of chemical herbicides. Twenty-five compounds were synthesized and tested. None of the compounds showed phytotoxic activity against C. benghalensis and C. sumatrensis, but almost all of them produced yellowing, bleaching, and necrosis on leaves of E. heterophylla. Two of the tyrosol 1,2,3-triazole derivatives produced more extensive lesions than those produced by the commercial herbicide diquat, used as a positive control (p ≤ 0.05). When applied on leaves of E. heterophylla, these compounds interfered with the stomatal conductance, net photosynthesis, internal carbon concentration, transpiration rate, water-use efficiency, and chlorophyll A and B contents. The interference of such compounds on such photosynthesis-related variables indicates that tyrosol 1,2,3-triazole derivatives may be capable of lowering the competitiveness of E. heterophylla and acting as additional tools for managing this competitive weed in agricultural lands.
Collapse
Affiliation(s)
- Cristiane Aparecida Franco
- Department of Chemistry, Universidade Federal de Viçosa, Av PH Rolfs sn, Viçosa, Minas Gerais 36.570-900, Brazil
| | - Toshik Iarley da Silva
- Department of Agronomy, Universidade Federal de Viçosa, Av PH Rolfs sn, Viçosa, Minas Gerais 36.570-900, Brazil
| | - Marlon Gomes Dias
- Department of Agronomy, Universidade Federal de Viçosa, Av PH Rolfs sn, Viçosa, Minas Gerais 36.570-900, Brazil
| | - Bruno Wesley Ferreira
- Department of Phytopathology, Universidade Federal de Viçosa, Av PH Rolfs sn, Viçosa, Minas Gerais 36.570-900, Brazil
| | - Bianca Lana de Sousa
- Department of Chemistry, Universidade Federal de Viçosa, Av PH Rolfs sn, Viçosa, Minas Gerais 36.570-900, Brazil
| | - Guilherme Mateus Bousada
- Department of Chemistry, Universidade Federal de Viçosa, Av PH Rolfs sn, Viçosa, Minas Gerais 36.570-900, Brazil
| | - Robert Weingart Barreto
- Department of Phytopathology, Universidade Federal de Viçosa, Av PH Rolfs sn, Viçosa, Minas Gerais 36.570-900, Brazil
| | - Boniek Gontijo Vaz
- Institute of Chemistry, Universidade Federal de Goiás, Av Esperança, sn, Samambaia, Goiânia 74.690-900, Brazil
| | - Gesiane da Silva Lima
- Institute of Chemistry, Universidade Federal de Goiás, Av Esperança, sn, Samambaia, Goiânia 74.690-900, Brazil
| | - Marcelo Henrique Dos Santos
- Department of Chemistry, Universidade Federal de Viçosa, Av PH Rolfs sn, Viçosa, Minas Gerais 36.570-900, Brazil
| | - José Antônio Saraiva Grossi
- Department of Agronomy, Universidade Federal de Viçosa, Av PH Rolfs sn, Viçosa, Minas Gerais 36.570-900, Brazil
| | | |
Collapse
|
13
|
Pandiri M, Nukala SK, Swamy TN, Dasari G, Badithapuram V, Manchal R, Bandari S. Design, Synthesis, and Anticancer Activity of Some New N-{5-[(1H-Benzo[d]imidazol-1-yl)methyl]isoxazol-3-yl}benzamide Hybrids. RUSS J GEN CHEM+ 2021. [DOI: 10.1134/s1070363221110207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
14
|
Subbaiah MAM, Meanwell NA. Bioisosteres of the Phenyl Ring: Recent Strategic Applications in Lead Optimization and Drug Design. J Med Chem 2021; 64:14046-14128. [PMID: 34591488 DOI: 10.1021/acs.jmedchem.1c01215] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The benzene moiety is the most prevalent ring system in marketed drugs, underscoring its historic popularity in drug design either as a pharmacophore or as a scaffold that projects pharmacophoric elements. However, introspective analyses of medicinal chemistry practices at the beginning of the 21st century highlighted the indiscriminate deployment of phenyl rings as an important contributor to the poor physicochemical properties of advanced molecules, which limited their prospects of being developed into effective drugs. This Perspective deliberates on the design and applications of bioisosteric replacements for a phenyl ring that have provided practical solutions to a range of developability problems frequently encountered in lead optimization campaigns. While the effect of phenyl ring replacements on compound properties is contextual in nature, bioisosteric substitution can lead to enhanced potency, solubility, and metabolic stability while reducing lipophilicity, plasma protein binding, phospholipidosis potential, and inhibition of cytochrome P450 enzymes and the hERG channel.
Collapse
Affiliation(s)
- Murugaiah A M Subbaiah
- Department of Medicinal Chemistry, Biocon-Bristol Myers Squibb Research and Development Centre, Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bangalore, Karnataka 560099, India
| | - Nicholas A Meanwell
- Department of Small Molecule Drug Discovery, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| |
Collapse
|
15
|
Svensen N, Wyllie S, Gray DW, De Rycker M. Live-imaging rate-of-kill compound profiling for Chagas disease drug discovery with a new automated high-content assay. PLoS Negl Trop Dis 2021; 15:e0009870. [PMID: 34634052 PMCID: PMC8530327 DOI: 10.1371/journal.pntd.0009870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/21/2021] [Accepted: 10/04/2021] [Indexed: 11/19/2022] Open
Abstract
Chagas disease, caused by the protozoan intracellular parasite Trypanosoma cruzi, is a highly neglected tropical disease, causing significant morbidity and mortality in central and south America. Current treatments are inadequate, and recent clinical trials of drugs inhibiting CYP51 have failed, exposing a lack of understanding of how to translate laboratory findings to the clinic. Following these failures many new model systems have been developed, both in vitro and in vivo, that provide improved understanding of the causes for clinical trial failures. Amongst these are in vitro rate-of-kill (RoK) assays that reveal how fast compounds kill intracellular parasites. Such assays have shown clear distinctions between the compounds that failed in clinical trials and the standard of care. However, the published RoK assays have some key drawbacks, including low time-resolution and inability to track the same cell population over time. Here, we present a new, live-imaging RoK assay for intracellular T. cruzi that overcomes these issues. We show that the assay is highly reproducible and report high time-resolution RoK data for key clinical compounds as well as new chemical entities. The data generated by this assay allow fast acting compounds to be prioritised for progression, the fate of individual parasites to be tracked, shifts of mode-of-action within series to be monitored, better PKPD modelling and selection of suitable partners for combination therapy. Chagas disease is caused by the single cell protozoan parasite Trypanosoma cruzi. Millions of people suffer from this disease in central and south America, which frequently causes heart disease and can result in death. Chagas disease is classified as a neglected tropical disease due to the lack of investment in development of new medicines. The currently available medicines are inadequate as they require long treatments, often with severe side-effects. To develop new medicines, it is critical to build laboratory assays and tools that help predict the ability of new compounds to cure patients. Rate-of-kill assays measure how quickly compounds can kill parasites, providing a route to differentiate promising compounds from poor ones. Here, we describe development of an advanced rate-of-kill assay that, unlike existing assays, can monitor the same cell population over the duration of compound treatment. Using live-cell microscopy, parasite-infected host cells and their response to compound treatment can be continuously monitored. This enables better defined rate-of-kill profiles to be produced, in turn allowing better informed decisions on subsequent compound progression. Here, we report the live-imaging rate-of-kill profiles for several key compounds, including current drugs and compounds in clinical development.
Collapse
Affiliation(s)
- Nina Svensen
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Susan Wyllie
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - David W Gray
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Manu De Rycker
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
16
|
Silva TB, Ji KNK, Petzold Pauli F, Galvão RMS, Faria AFM, Bello ML, Resende JALC, Campos VR, Forezi LDSM, da Silva FDC, Faria RX, Ferreira VF. Synthesis and in vitro and in silico studies of 1H- and 2H-1,2,3-triazoles as antichagasic agents. Bioorg Chem 2021; 116:105250. [PMID: 34469833 DOI: 10.1016/j.bioorg.2021.105250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/13/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
1,2,3-triazole heterocycles stand out in medicinal chemistry for having great structural diversity and bioactivities. In this study, two series of triazoles were synthesized. One was obtained by the 1,3-dipolar cycloaddition reaction between ethyl cyanoacetate and several phenyl azides forming 1H-1,2,3-triazoles and the other by rearrangement of Dimroth forming and 2H-1,2,3-triazoles. Both series were shown to be active against the epimastigote form of Trypanosoma cruzi. The 1,2,3-triazoles 16d (S.I. between 100 and 200), 17d and 16f (S.I. > 200) were the most active compounds and capable of breaking the plasma membrane of trypomastigotes acting on CYP51 and inhibiting ergosterol synthesis. Candidate 16d exhibited the best and most favorable profile when interacting with CYP51.
Collapse
Affiliation(s)
- Thais B Silva
- Universidade Federal Fluminense, Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Santa Rosa, CEP 24241-002 Niterói, RJ, Brazil
| | - Kathya N K Ji
- Universidade Federal Fluminense, Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Santa Rosa, CEP 24241-002 Niterói, RJ, Brazil
| | - Fernanda Petzold Pauli
- Universidade Federal Fluminense, Instituto de Química, Departamento de Química Orgânica, Campus do Valonguinho, CEP 24020-150 Niterói, RJ, Brazil
| | - Raíssa M S Galvão
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Laboratório de Toxoplasmose e outras Protozooses, Pavilhão Carlos Chagas, Manguinhos, CEP 21045-900 Rio de Janeiro, RJ, Brazil; Universidade Federal Fluminense, Instituto de Biologia, Pós-graduação de Ciências e Biotecnologia, Campus do Valonguinho, CEP 24020-150 Niterói, RJ, Brazil
| | - Ana F M Faria
- Universidade Federal do Rio de Janeiro, Faculdade de Farmácia, Laboratório de Planejamento Farmacêutico e Simulação Computacional, CEP 21941-599 Rio de Janeiro, RJ, Brazil
| | - Murilo L Bello
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Laboratório de Toxoplasmose e outras Protozooses, Pavilhão Carlos Chagas, Manguinhos, CEP 21045-900 Rio de Janeiro, RJ, Brazil
| | - Jackson A L C Resende
- Universidade Federal de Mato Grosso, Instituto de Ciências Exatas e da Terra, Campus Universitário do Araguaia, CEP 78698-000 Pontal do Araguaia, MT, Brazil
| | - Vinicius R Campos
- Universidade Federal Fluminense, Instituto de Química, Departamento de Química Orgânica, Campus do Valonguinho, CEP 24020-150 Niterói, RJ, Brazil
| | - Luana da S M Forezi
- Universidade Federal Fluminense, Instituto de Química, Departamento de Química Orgânica, Campus do Valonguinho, CEP 24020-150 Niterói, RJ, Brazil
| | - Fernando de C da Silva
- Universidade Federal Fluminense, Instituto de Química, Departamento de Química Orgânica, Campus do Valonguinho, CEP 24020-150 Niterói, RJ, Brazil
| | - Robson X Faria
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Laboratório de Toxoplasmose e outras Protozooses, Pavilhão Carlos Chagas, Manguinhos, CEP 21045-900 Rio de Janeiro, RJ, Brazil; Universidade Federal do Rio de Janeiro, Faculdade de Farmácia, Laboratório de Planejamento Farmacêutico e Simulação Computacional, CEP 21941-599 Rio de Janeiro, RJ, Brazil.
| | - Vitor F Ferreira
- Universidade Federal Fluminense, Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Santa Rosa, CEP 24241-002 Niterói, RJ, Brazil.
| |
Collapse
|
17
|
Taylor MC, Ward AI, Olmo F, Francisco AF, Jayawardhana S, Costa FC, Lewis MD, Kelly JM. Bioluminescent:Fluorescent Trypanosoma cruzi Reporter Strains as Tools for Exploring Chagas Disease Pathogenesis and Drug Activity. Curr Pharm Des 2021; 27:1733-1740. [PMID: 33234096 DOI: 10.2174/1381612826666201124113214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/16/2020] [Accepted: 07/23/2020] [Indexed: 11/22/2022]
Abstract
Chagas disease results from infection with the trypanosomatid parasite Trypanosoma cruzi. Progress in developing new drugs has been hampered by the long term and complex nature of the condition and by our limited understanding of parasite biology. Technical difficulties in assessing the parasite burden during the chronic stage of infection have also proven to be a particular challenge. In this context, the development of noninvasive, highly sensitive bioluminescence imaging procedures based on parasites that express a red-shifted luciferase has greatly enhanced our ability to monitor infections in experimental models. Applications of this methodology have led to new insights into tissue tropism and infection dynamics and have been a major driver in drug development. The system has been further modified by the generation of parasite reporter lines that express bioluminescent:fluorescent fusion proteins, an advancement that has allowed chronic infections in mice to be examined at a cellular level. By exploiting bioluminescence, to identify the rare sites of tissue infection, and fluorescence to detect T. cruzi at the level of individual host cells in histological sections, it has been possible to investigate the replication and differentiation status of parasites in vivo and to examine the cellular environment of infection foci. In combination, these data provide a framework for the detailed dissection of disease pathogenesis and drug activity.
Collapse
Affiliation(s)
- Martin C Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Alexander I Ward
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Amanda F Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Fernanda C Costa
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Michael D Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - John M Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| |
Collapse
|
18
|
Grimberg DC, Shah A, Tan WP, Etienne W, Spasojevic I, Inman BA. Hyperthermia Improves Solubility of Intravesical Chemotherapeutic Agents. Bladder Cancer 2020; 6:461-470. [PMID: 36118287 PMCID: PMC9441059 DOI: 10.3233/blc-200350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/24/2020] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Nearly 70% of all new cases of bladder cancer are non-muscle invasive disease, the treatment for which includes transurethral resection followed by intravesical therapy. Unfortunately, recurrence rates approach 50% in part due to poor intravesical drug delivery. Hyperthermia is frequently used as an adjunct to intravesical chemotherapy to improve drug delivery and response to treatment. OBJECTIVE: To assess the solubility profile of intravesical chemotherapies under varying conditions of pH and temperature. METHODS: Using microplate laser nephelometry we measured the solubility of three intravesical chemotherapy agents (mitomycin C, gemcitabine, and cisplatin) at varying physical conditions. Drugs were assessed at room temperature (23°C), body temperature (37°C), and 43°C, the temperature used for hyperthermic intravesical treatments. To account for variations in urine pH, solubility was also investigated at pH 4.00, 6.00, and 8.00. RESULTS: Heat incrementally increased the solubility of all three drugs studied. Conversely, pH largely did not impact solubility aside for gemcitabine which showed slightly reduced solubility at pH 8.00 versus 6.00 or 4.00. Mitomycin C at the commonly used 2.0 mg/mL was insoluble at room temperature, but soluble at both 37 and 43°C. CONCLUSIONS: Hyperthermia as an adjunct to intravesical treatment would improve drug solubility, and likely drug delivery as some current regimens are insoluble without heat. Improvements in solubility also allow for testing of alternative administration regimens to improve drug delivery or tolerability. Further studies are needed to confirm that improvements in solubility result in increased drug delivery.
Collapse
Affiliation(s)
- Dominic C. Grimberg
- Division of Urology, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Ankeet Shah
- Division of Urology, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Wei Phin Tan
- Division of Urology, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Wiguins Etienne
- Division of Urology, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Ivan Spasojevic
- Department of Medicine, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Brant A. Inman
- Division of Urology, Duke Prostate and Urologic Cancer Center, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
19
|
Selected 5-amino-1-aryl-1H-1,2,3-triazole scaffolds as promising antiproliferative agents. UKRAINIAN BIOCHEMICAL JOURNAL 2020. [DOI: 10.15407/ubj92.05.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
20
|
Scarim CB, Chin CM. Current Approaches to Drug Discovery for Chagas Disease: Methodological Advances. Comb Chem High Throughput Screen 2020; 22:509-520. [PMID: 31608837 DOI: 10.2174/1386207322666191010144111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/31/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND In recent years, there has been an improvement in the in vitro and in vivo methodology for the screening of anti-chagasic compounds. Millions of compounds can now have their activity evaluated (in large compound libraries) by means of high throughput in vitro screening assays. OBJECTIVE Current approaches to drug discovery for Chagas disease. METHOD This review article examines the contribution of these methodological advances in medicinal chemistry in the last four years, focusing on Trypanosoma cruzi infection, obtained from the PubMed, Web of Science, and Scopus databases. RESULTS Here, we have shown that the promise is increasing each year for more lead compounds for the development of a new drug against Chagas disease. CONCLUSION There is increased optimism among those working with the objective to find new drug candidates for optimal treatments against Chagas disease.
Collapse
Affiliation(s)
- Cauê B Scarim
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, Brazil.,Lapdesf - Laboratory of Research and Development of Drugs, Araraquara, São Paulo, Brazil
| | - Chung M Chin
- Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, Brazil.,Lapdesf - Laboratory of Research and Development of Drugs, Araraquara, São Paulo, Brazil
| |
Collapse
|
21
|
Thomas MG, De Rycker M, Wall RJ, Spinks D, Epemolu O, Manthri S, Norval S, Osuna-Cabello M, Patterson S, Riley J, Simeons FRC, Stojanovski L, Thomas J, Thompson S, Naylor C, Fiandor JM, Wyatt PG, Marco M, Wyllie S, Read KD, Miles TJ, Gilbert IH. Identification and Optimization of a Series of 8-Hydroxy Naphthyridines with Potent In Vitro Antileishmanial Activity: Initial SAR and Assessment of In Vivo Activity. J Med Chem 2020; 63:9523-9539. [PMID: 32663005 PMCID: PMC7748245 DOI: 10.1021/acs.jmedchem.0c00705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
![]()
Visceral
leishmaniasis (VL) is a parasitic infection that results
in approximately 26 000–65 000 deaths annually.
The available treatments are hampered by issues such as toxicity,
variable efficacy, and unsuitable dosing options. The need for new
treatments is urgent and led to a collaboration between the Drugs
for Neglected Diseases initiative (DNDi), GlaxoSmithKline (GSK), and the University of Dundee. An 8-hydroxynaphthyridine
was identified as a start point, and an early compound demonstrated
weak efficacy in a mouse model of VL but was hampered by glucuronidation.
Efforts to address this led to the development of compounds with improved in vitro profiles, but these were poorly tolerated in vivo. Investigation of the mode of action (MoA) demonstrated
that activity was driven by sequestration of divalent metal cations,
a mechanism which was likely to drive the poor tolerability. This
highlights the importance of investigating MoA and pharmacokinetics
at an early stage for phenotypically active series.
Collapse
Affiliation(s)
- Michael G Thomas
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Manu De Rycker
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Richard J Wall
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Daniel Spinks
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Ola Epemolu
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Sujatha Manthri
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Suzanne Norval
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Maria Osuna-Cabello
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Stephen Patterson
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Jennifer Riley
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Frederick R C Simeons
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Laste Stojanovski
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - John Thomas
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Stephen Thompson
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Claire Naylor
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Jose M Fiandor
- Global Health R&D, GlaxoSmithKline, Tres Cantos 28760, Spain
| | - Paul G Wyatt
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Maria Marco
- Global Health R&D, GlaxoSmithKline, Tres Cantos 28760, Spain
| | - Susan Wyllie
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Kevin D Read
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Timothy J Miles
- Global Health R&D, GlaxoSmithKline, Tres Cantos 28760, Spain
| | - Ian H Gilbert
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| |
Collapse
|
22
|
Francisco AF, Jayawardhana S, Olmo F, Lewis MD, Wilkinson SR, Taylor MC, Kelly JM. Challenges in Chagas Disease Drug Development. Molecules 2020; 25:E2799. [PMID: 32560454 PMCID: PMC7355550 DOI: 10.3390/molecules25122799] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 02/08/2023] Open
Abstract
The protozoan parasite Trypanosoma cruzi causes Chagas disease, an important public health problem throughout Latin America. Current therapeutic options are characterised by limited efficacy, long treatment regimens and frequent toxic side-effects. Advances in this area have been compromised by gaps in our knowledge of disease pathogenesis, parasite biology and drug activity. Nevertheless, several factors have come together to create a more optimistic scenario. Drug-based research has become more systematic, with increased collaborations between the academic and commercial sectors, often within the framework of not-for-profit consortia. High-throughput screening of compound libraries is being widely applied, and new technical advances are helping to streamline the drug development pipeline. In addition, drug repurposing and optimisation of current treatment regimens, informed by laboratory research, are providing a basis for new clinical trials. Here, we will provide an overview of the current status of Chagas disease drug development, highlight those areas where progress can be expected, and describe how fundamental research is helping to underpin the process.
Collapse
Affiliation(s)
- Amanda F. Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - Shane R. Wilkinson
- School of Biological and Chemical Sciences, Queen Mary University of London Mile End Road, London E1 4NS, UK;
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine Keppel Street, London WC1E 7HT, UK; (A.F.F.); (S.J.); (F.O.); (M.D.L.); (M.C.T.)
| |
Collapse
|
23
|
Tupychak MA, Shyyka OY, Pokhodylo NT, Obushak MD. Nitrileimines as an alternative to azides in base-mediated click [3 + 2] cycloaddition with methylene active nitriles. RSC Adv 2020; 10:13696-13699. [PMID: 35493004 PMCID: PMC9051556 DOI: 10.1039/d0ra01417f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 03/20/2020] [Indexed: 12/30/2022] Open
Abstract
Nitrileimines were implemented in practical click protocols with oxopropanenitriles for the straightforward 5-amino-1H-pyrazole synthesis via 1,3-dipolar cycloaddition. The reaction proceeds at room temperature in a short time with base catalysis and no chromatographic purification. High purity products were isolated by simple filtration. The selectivity of the reaction was observed.
Collapse
Affiliation(s)
- Mykola A Tupychak
- Department of Organic Chemistry, Ivan Franko National University of Lviv Kyryla i Mefodiya St. 6 Lviv 79005 Ukraine
| | - Olga Ya Shyyka
- Department of Organic Chemistry, Ivan Franko National University of Lviv Kyryla i Mefodiya St. 6 Lviv 79005 Ukraine
| | - Nazariy T Pokhodylo
- Department of Organic Chemistry, Ivan Franko National University of Lviv Kyryla i Mefodiya St. 6 Lviv 79005 Ukraine
| | - Mykola D Obushak
- Department of Organic Chemistry, Ivan Franko National University of Lviv Kyryla i Mefodiya St. 6 Lviv 79005 Ukraine
| |
Collapse
|
24
|
Wall RJ, Carvalho S, Milne R, Bueren-Calabuig JA, Moniz S, Cantizani-Perez J, MacLean L, Kessler A, Cotillo I, Sastry L, Manthri S, Patterson S, Zuccotto F, Thompson S, Martin J, Marco M, Miles TJ, De Rycker M, Thomas MG, Fairlamb AH, Gilbert IH, Wyllie S. The Q i Site of Cytochrome b is a Promiscuous Drug Target in Trypanosoma cruzi and Leishmania donovani. ACS Infect Dis 2020; 6:515-528. [PMID: 31967783 PMCID: PMC7076694 DOI: 10.1021/acsinfecdis.9b00426] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Indexed: 01/29/2023]
Abstract
Available treatments for Chagas' disease and visceral leishmaniasis are inadequate, and there is a pressing need for new therapeutics. Drug discovery efforts for both diseases principally rely upon phenotypic screening. However, the optimization of phenotypically active compounds is hindered by a lack of information regarding their molecular target(s). To combat this issue we initiate target deconvolution studies at an early stage. Here, we describe comprehensive genetic and biochemical studies to determine the targets of three unrelated phenotypically active compounds. All three structurally diverse compounds target the Qi active-site of cytochrome b, part of the cytochrome bc1 complex of the electron transport chain. Our studies go on to identify the Qi site as a promiscuous drug target in Leishmania donovani and Trypanosoma cruzi with a propensity to rapidly mutate. Strategies to rapidly identify compounds acting via this mechanism are discussed to ensure that drug discovery portfolios are not overwhelmed with inhibitors of a single target.
Collapse
Affiliation(s)
- Richard J. Wall
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Sandra Carvalho
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Rachel Milne
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Juan A. Bueren-Calabuig
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Sonia Moniz
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | | | - Lorna MacLean
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Albane Kessler
- Global Health R&D, GlaxoSmithKline, Tres Cantos 28760, Spain
| | - Ignacio Cotillo
- Global Health R&D, GlaxoSmithKline, Tres Cantos 28760, Spain
| | - Lalitha Sastry
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Sujatha Manthri
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Stephen Patterson
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Fabio Zuccotto
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Stephen Thompson
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Julio Martin
- Global Health R&D, GlaxoSmithKline, Tres Cantos 28760, Spain
| | - Maria Marco
- Global Health R&D, GlaxoSmithKline, Tres Cantos 28760, Spain
| | | | - Manu De Rycker
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Michael G. Thomas
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Alan H. Fairlamb
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| | - Ian H. Gilbert
- Drug Discovery Unit,
Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Susan Wyllie
- Division of Biological
Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives
Research, School of Life Sciences, University
of Dundee, Dow Street, Dundee DD1
5EH, United Kingdom
| |
Collapse
|
25
|
Lemport PS, Roznyatovsky VA, Tarasevich BN, Khromov OV, Khrustalev VN, Rozentsveig IB, Nenajdenko VG. Reaction of 3-azidoisoxazoles with active methylene compounds. MENDELEEV COMMUNICATIONS 2019. [DOI: 10.1016/j.mencom.2019.09.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
26
|
Flow Hydrodediazoniation of Aromatic Heterocycles. MOLECULES (BASEL, SWITZERLAND) 2019; 24:molecules24101996. [PMID: 31137676 PMCID: PMC6572451 DOI: 10.3390/molecules24101996] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 12/31/2022]
Abstract
Continuous flow processing was applied for the rapid replacement of an aromatic amino group with a hydride. The approach was applied to a range of aromatic heterocycles, confirming the wide scope and substituent-tolerance of the processes. Flow equipment was utilized and the process optimised to overcome the problematically-unstable intermediates that have restricted yields in previous studies relying on batch procedures. Various common organic solvents were investigated as potential hydride sources. The approach has allowed key structures, such as amino-pyrazoles and aminopyridines, to be deaminated in good yield using a purely organic-soluble system.
Collapse
|
27
|
Hulpia F, Van Hecke K, França da Silva C, da Gama Jaen Batista D, Maes L, Caljon G, de Nazaré C Soeiro M, Van Calenbergh S. Discovery of Novel 7-Aryl 7-Deazapurine 3'-Deoxy-ribofuranosyl Nucleosides with Potent Activity against Trypanosoma cruzi. J Med Chem 2018; 61:9287-9300. [PMID: 30234983 DOI: 10.1021/acs.jmedchem.8b00999] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chagas disease is the leading cause of cardiac-related mortality in Latin American countries where it is endemic. Trypanosoma cruzi, the disease-causing pathogen, is unable to synthesize purines de novo, necessitating salvage of preformed host purines. Therefore, purine and purine-nucleoside analogues might constitute an attractive source for identifying antitrypanosomal hits. In this study, structural elements of two purine-nucleoside analogues (i.e., cordycepin and a recently discovered 7-substituted 7-deazaadenosine) led to the identification of novel nucleoside analogues with potent in vitro activity. The structure-activity relationships of substituents at C-7 were investigated, ultimately leading to the selection of compound 5, with a C-7 para-chlorophenyl group, for in vivo evaluation. This derivative showed complete suppression of T. cruzi Y-strain blood parasitemia when orally administered twice daily for 5 days at 25 mg/kg and was able to protect infected mice from parasite-induced mortality. However, sterile cure by immunosuppression could not be demonstrated.
Collapse
Affiliation(s)
- Fabian Hulpia
- Laboratory for Medicinal Chemistry (Campus Heymans) , Ghent University , Ottergemsesteenweg 460 , Gent B-9000 , Belgium
| | - Kristof Van Hecke
- XStruct, Department of Chemistry , Ghent University , Krijgslaan 281 S3 , Gent B-9000 , Belgium
| | - Cristiane França da Silva
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz (FIOCRUZ) , Fundação Oswaldo Cruz , Avenida Brasil, 4365 , Manguinhos, Rio de Janeiro , RJ 21040-900 , Brazil
| | - Denise da Gama Jaen Batista
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz (FIOCRUZ) , Fundação Oswaldo Cruz , Avenida Brasil, 4365 , Manguinhos, Rio de Janeiro , RJ 21040-900 , Brazil
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene , University of Antwerp , Universiteitsplein 1 (S7) , Wilrijk B-2610 , Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene , University of Antwerp , Universiteitsplein 1 (S7) , Wilrijk B-2610 , Belgium
| | - Maria de Nazaré C Soeiro
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz (FIOCRUZ) , Fundação Oswaldo Cruz , Avenida Brasil, 4365 , Manguinhos, Rio de Janeiro , RJ 21040-900 , Brazil
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry (Campus Heymans) , Ghent University , Ottergemsesteenweg 460 , Gent B-9000 , Belgium
| |
Collapse
|
28
|
Li L, Xing X, Zhang C, Zhu A, Fan X, Chen C, Zhang G. Novel synthesis of 5-iodo-1,2,3-triazoles using an aqueous iodination system under air. Tetrahedron Lett 2018. [DOI: 10.1016/j.tetlet.2018.08.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
29
|
Thomas MG, De Rycker M, Cotillo Torrejon I, Thomas J, Riley J, Spinks D, Read KD, Miles TJ, Gilbert IH, Wyatt PG. 2,4-Diamino-6-methylpyrimidines for the potential treatment of Chagas' disease. Bioorg Med Chem Lett 2018; 28:3025-3030. [PMID: 30104093 DOI: 10.1016/j.bmcl.2018.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/25/2018] [Accepted: 08/03/2018] [Indexed: 01/15/2023]
Abstract
Chagas' disease, caused by the protozoan parasite Trypanosoma cruzi, affects 8-10 million people across the Latin American population and is responsible for around 12,500 deaths per annum. The current frontline treatments, benznidazole and nifurtimox, are associated with side effects and lack efficacy in the chronic stage of the disease, leading to an urgent need for new treatments. A high throughput screening campaign against the physiologically relevant intracellular form of the parasite identified a series of 2,4-diamino-6-methylpyrimidines. Demonstrating the series did not work through the anti-target TcCYP51, and was generally cytocidal, confirmed its suitability for further development. This study reports the optimisation of selectivity and metabolic stability of the series and identification of a suitable lead for further optimisation.
Collapse
Affiliation(s)
- Michael G Thomas
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Manu De Rycker
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Ignacio Cotillo Torrejon
- Kinetoplastid DPU, Global Health R&D, Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Spain
| | - John Thomas
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Jennifer Riley
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Daniel Spinks
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Kevin D Read
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Tim J Miles
- Kinetoplastid DPU, Global Health R&D, Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Spain
| | - Ian H Gilbert
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Paul G Wyatt
- Drug Discovery Unit, Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom.
| |
Collapse
|
30
|
Peres RB, Ullah AI, de Almeida Fiuza LF, Silva PB, Batista MM, Corcoran O, Reddy TRK, de Nazaré Correia Soeiro M. Identification and preliminary structure-activity relationship studies of novel pyridyl sulfonamides as potential Chagas disease therapeutic agents. Bioorg Med Chem Lett 2018; 28:2018-2022. [PMID: 29748049 DOI: 10.1016/j.bmcl.2018.04.064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/25/2018] [Accepted: 04/27/2018] [Indexed: 10/17/2022]
Abstract
Chagas disease is a neglected pathology responsible for about 12,000 deaths every year across Latin America. Although six million people are infected by the Trypanosoma cruzi, current therapeutic options are limited, highlighting the need for new drugs. Here we report the preliminary structure activity relationships of a small library of 17 novel pyridyl sulfonamide derivatives. Analogues 4 and 15 displayed significant potency against intracellular amastigotes with EC50 of 5.4 µM and 8.6 µM. In cytotoxicity assays using mice fibroblast L929 cell lines, both compounds indicated low toxicity with decent selectivity indices (SI) >36 and >23 respectively. Hence these compounds represent good starting points for further lead optimization.
Collapse
Affiliation(s)
- Raiza Brandão Peres
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Asma Inam Ullah
- The Medicines Research Group, School of Health, Sport and Bioscience, College of Applied Health and Communities, University of East London, Stratford Campus, Water Lane, E15 4LZ, UK
| | | | - Patricia Bernardino Silva
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Marcos M Batista
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Olivia Corcoran
- The Medicines Research Group, School of Health, Sport and Bioscience, College of Applied Health and Communities, University of East London, Stratford Campus, Water Lane, E15 4LZ, UK
| | - Tummala Rama Krishna Reddy
- The Medicines Research Group, School of Health, Sport and Bioscience, College of Applied Health and Communities, University of East London, Stratford Campus, Water Lane, E15 4LZ, UK.
| | | |
Collapse
|
31
|
Fan YL, Jin XH, Huang ZP, Yu HF, Zeng ZG, Gao T, Feng LS. Recent advances of imidazole-containing derivatives as anti-tubercular agents. Eur J Med Chem 2018; 150:347-365. [PMID: 29544148 DOI: 10.1016/j.ejmech.2018.03.016] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/02/2018] [Accepted: 03/04/2018] [Indexed: 12/20/2022]
Abstract
Tuberculosis still remains one of the most common, communicable, and leading deadliest diseases known to mankind throughout the world. Drug-resistance in Mycobacterium tuberculosis which threatens to worsen the global tuberculosis epidemic has caused great concern in recent years. To overcome the resistance, the development of new drugs with novel mechanisms of actions is of great importance. Imidazole-containing derivatives endow with various biological properties, and some of them demonstrated excellent anti-tubercular activity. As the most emblematic example, 4-nitroimidazole delamanid has already received approval for treatment of multidrug-resistant tuberculosis infected patients. Thus, imidazole-containing derivatives have caused great interests in discovery of new anti-tubercular agents. Numerous of imidazole-containing derivatives were synthesized and screened for their in vitro and in vivo anti-mycobacterial activities against both drug-sensitive and drug-resistant Mycobacterium tuberculosis pathogens. This review aims to outline the recent advances of imidazole-containing derivatives as anti-tubercular agents, and summarize the structure-activity relationship of these derivatives. The enriched structure-activity relationship may pave the way for the further rational development of imidazole-containing derivatives as anti-tubercular agents.
Collapse
Affiliation(s)
- Yi-Lei Fan
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, Zhejiang Police College, Hangzhou, PR China
| | - Xiao-Hong Jin
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Zhong-Ping Huang
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou, PR China.
| | - Hai-Feng Yu
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Zhi-Gang Zeng
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Tao Gao
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning 437100, PR China.
| | - Lian-Shun Feng
- Synthetic and Functional Biomolecules Center, Peking University, Beijing, PR China
| |
Collapse
|