1
|
da Silva Bortoleti BT, Camargo PG, Gonçalves MD, Tomiotto-Pellissier F, Silva TF, Concato VM, Detoni MB, Bidóia DL, da Silva Lima CH, Rodrigues CR, Bispo MDLF, de Macedo FC, Conchon-Costa I, Miranda-Sapla MM, Wowk PF, Pavanelli WR. Effect of a thiohydantoin salt derived from l-Arginine on Leishmania amazonensis and infected cells: Insights from biological effects to molecular docking interactions. Chem Biol Interact 2024; 403:111216. [PMID: 39218371 DOI: 10.1016/j.cbi.2024.111216] [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] [Revised: 07/12/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Leishmaniasis is a neglected tropical disease caused by parasites of the genus Leishmania and is responsible for more than 1 million new cases and 70,000 deaths annually worldwide. Treatment has high costs, toxicity, complex and long administration time, several adverse effects, and drug-resistant strains, therefore new therapies are urgently needed. Synthetic compounds have been highlighted in the medicinal chemistry field as a strong option for drug development against different diseases. Organic salts (OS) have multiple biological activities, including activity against protozoa such as Leishmania spp. This study aimed to investigate the in vitro leishmanicidal activity and death mechanisms of a thiohydantoin salt derived from l-arginine (ThS) against Leishmania amazonensis. We observed that ThS treatment inhibited promastigote proliferation, increased ROS production, phosphatidylserine exposure and plasma membrane permeabilization, loss of mitochondrial membrane potential, lipid body accumulation, autophagic vacuole formation, cell cycle alteration, and morphological and ultrastructural changes, showing parasites death. Additionally, ThS presents low cytotoxicity in murine macrophages (J774A.1), human monocytes (THP-1), and sheep erythrocytes. ThS in vitro cell treatment reduced the percentage of infected macrophages and the number of amastigotes per macrophage by increasing ROS production and reducing TNF-α levels. These results highlight the potential of ThS among thiohydantoins, mainly related to the arginine portion, as a leishmanicidal drug for future drug strategies for leishmaniasis treatment. Notably, in silico investigation of key targets from L. amazonensis, revealed that a ThS compound from the l-arginine amino acid strongly interacts with arginase (ARG) and TNF-α converting enzyme (TACE), suggesting its potential as a Leishmania inhibitor.
Collapse
Affiliation(s)
- Bruna Taciane da Silva Bortoleti
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute, (ICC/Fiocruz/PR), Curitiba, Paraná, Brazil; State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Priscila Goes Camargo
- Federal University of Rio de Janeiro, Faculty of Pharmacy, Rio de Janeiro, Rio de Janeiro, Brazil; State University of Londrina (UEL/PR), Chemistry Department, Londrina, Paraná, Brazil
| | - Manoela Daiele Gonçalves
- State University of Londrina (UEL/PR), Laboratory of Biotransformation and Phytochemistry, Londrina, Paraná, Brazil
| | - Fernanda Tomiotto-Pellissier
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute, (ICC/Fiocruz/PR), Curitiba, Paraná, Brazil; State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Taylon Felipe Silva
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Virginia Marcia Concato
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Mariana Barbosa Detoni
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | - Danielle Larazin Bidóia
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | | | - Carlos Rangel Rodrigues
- Federal University of Rio de Janeiro, Faculty of Pharmacy, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Ivete Conchon-Costa
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil
| | | | - Pryscilla Fanini Wowk
- Carlos Chagas Institute (ICC/Fiocruz/PR), Molecular Immunology and Cellular Group, Curitiba, Paraná, Brazil.
| | - Wander Rogério Pavanelli
- State University of Londrina (UEL/PR), Laboratory of Immunoparasitology, Londrina, Paraná, Brazil.
| |
Collapse
|
2
|
Kawato K, Sato K, Wada T. α-Selective Solid-Phase Synthesis of Glycosyl Phosphate Repeating Structure Via the Phosphoramidite Method. Chemistry 2024; 30:e202401226. [PMID: 39023079 DOI: 10.1002/chem.202401226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Lipophosphoglycans (LPGs) are found on the surface of Leishmania, a protozoan parasite, and are immunologically important. Herein, disaccharide 1-phosphate repeating units of LPGs were successfully synthesized on a solid support with high anomeric purity using a disaccharide α-1-phosphoramidite building block. To enhance solubility in the reaction solvent, hydroxy-protecting groups in the form of para-t-butylbenzoyl were introduced to the building block. The saccharide chain was elongated via stable glycosyl boranophosphate linkages, followed by the conversion of inter-sugar linkages to phosphodiester counterparts using an oxaziridine derivative. The addition of a silylating reagent post-reaction with the oxaziridine derivative efficiently facilitated the conversion of boranophosohodiesters to phosphodiesters. This method enabled the α-selective synthesis of up to 15 repeating units, marking the longest homogeneous repeating units of LPGs synthesized chemically. Given the chain length equivalence to native LPGs, the method developed herein holds promise for advancing anti-Leishmanial pharmaceuticals and vaccines.
Collapse
Affiliation(s)
- Kazuki Kawato
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda Chiba, 278-8510, Japan
| | - Kazuki Sato
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda Chiba, 278-8510, Japan
| | - Takeshi Wada
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda Chiba, 278-8510, Japan
| |
Collapse
|
3
|
Ribeiro JM, Teixeira EDM, Alves LL, Alves ÉAR, Pascoal-Xavier MA, Santi AMM, Oliveira E, Guimarães PPG, Teixeira-Carvalho A, Murta SMF, Peruhype-Magalhães V, Souza-Fagundes EM. Can letrozole be repurposed for the treatment of visceral leishmaniasis? Antimicrob Agents Chemother 2024:e0075624. [PMID: 39387580 DOI: 10.1128/aac.00756-24] [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: 06/17/2024] [Accepted: 09/06/2024] [Indexed: 10/15/2024] Open
Abstract
Visceral leishmaniasis, caused by Leishmania infantum in New World countries, is the most serious and potentially fatal form of leishmaniasis, if left untreated. There are currently no effective prophylactic measures, and therapeutic options are limited. Therefore, we investigated whether the aromatase inhibitor letrozole (LET), which is already used to treat breast cancer, has an antileishmanial activity and/or immunomodulatory potential and therefore may be used to treat L. infantum infection. LET was active against L. infantum promastigote and amastigote life cycle stages in an in vitro infection model using human THP-1 cell-derived macrophages. In human peripheral blood leukocytes ex vivo, LET reduced the internalized forms of L. infantum by classical monocytes and activated neutrophils. Concomitantly, LET stimulated the production of IL-12/TNF-α and decreased the production of IL-10/TGF-β by peripheral blood phagocytes, while in T and B cells, it promoted the production of TNF-α/IFN-γ and decreased that of IL-10. In a murine infection model, LET significantly reduced the parasite load in the liver after just 5 days and in the spleen after 15 days. During in vivo treatment with LET, the production of TNF-α/IFN-γ also increased. In addition, the proportion of developing granulomas decreased and that of mature granulomas increased in the liver, while there was no significant change in organ architecture in the spleen. Based on these data, repositioning of LET may be promising for the treatment of visceral leishmaniasis in humans.
Collapse
Affiliation(s)
- Juliana Martins Ribeiro
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Grupo de Genômica Funcional de Parasitos, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Eliane de Morais Teixeira
- Grupo Pesquisa Clínica e Políticas Públicas em Doenças Infecto-Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Líndicy Leidicy Alves
- Grupo Pesquisa Clínica e Políticas Públicas em Doenças Infecto-Parasitárias, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Érica Alessandra Rocha Alves
- Grupo Imunologia Celular e Molecular, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | | | - Ana Maria Murta Santi
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Edward Oliveira
- Grupo de Genômica Funcional de Parasitos, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro Pires Goulart Guimarães
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andrea Teixeira-Carvalho
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Silvane Maria Fonseca Murta
- Grupo de Genômica Funcional de Parasitos, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Vanessa Peruhype-Magalhães
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Elaine Maria Souza-Fagundes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
4
|
Shahabuddin, Uzma, Azam M, Parveen M, Kadir NHA, Min K, Alam M. Exploring 7β-amino-6-nitrocholestens as COVID-19 antivirals: in silico, synthesis, evaluation, and integration of artificial intelligence (AI) in drug design: assessing the cytotoxicity and antioxidant activity of 3β-acetoxynitrocholestane. RSC Med Chem 2024:d4md00257a. [PMID: 39430952 PMCID: PMC11485945 DOI: 10.1039/d4md00257a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 09/22/2024] [Indexed: 10/22/2024] Open
Abstract
In light of the ongoing pandemic caused by SARS-CoV-2, effective and clinically translatable treatments are desperately needed for COVID-19 and its emerging variants. In this study, some derivatives, including 7β-aminocholestene compounds, and 3β-acetoxy-6-nitrocholesta-4,6-diene were synthesized, in quantitative yields from 7β-bromo-6-nitrocholest-5-enes (1-3) with a small library of amines. The synthesized steroidal products were then thoroughly characterized using a range of physicochemical techniques, including IR, NMR, UV, MS, and elemental analysis. Next, a virtual screening based on structures using docking studies was conducted to investigate the potential of these synthesized compounds as therapeutic candidates against SARS-CoV-2. Specifically, we evaluated the compounds' binding energy of the reactants and their products with three SARS-CoV-2 functional proteins: the papain-like protease, 3C-like protease or main protease, and RNA-dependent RNA polymerase. Our results indicate that the 7β-aminocholestene derivatives (4-8) display intermediate to excellent binding energy, suggesting that they interact strongly with the receptor's active amino acids and may be promising drug candidates for inhibiting SARS-CoV-2. Although the starting steroid derivatives; 7β-bromo-6-nitrocholest-5-enes (1-3) and one steroid product; 3β-acetoxy-6-nitrocholesta-4,6-diene (9) exhibited strong binding energies with various SARS-CoV-2 receptors, they did not meet the Lipinski Rule and ADMET properties required for drug development. These compounds showed either mutagenic or reproductive/developmental toxicity when assessed using toxicity prediction software. The findings based on structure-based virtual screening, suggest that 7β-aminocholestaines (4-8) may be useful for reducing the susceptibility to SARS-CoV-2 infection. The docking pose of compound 4, which has a high score of -7.4 kcal mol-1, was subjected to AI-assisted deep learning to generate 60 AI-designed molecules for drug design. Molecular docking of these AI molecules was performed to select optimal candidates for further analysis and visualization. The cytotoxicity and antioxidant effects of 3β-acetoxy-6-nitrocholesta-4,6-diene were tested in vitro, showing marked cytotoxicity and antioxidant activity. To elucidate the molecular basis for these effects, steroidal compound 9 was subjected to molecular docking analysis to identify potential binding interactions. The stability of the top-ranked docking pose was subsequently assessed using molecular dynamics simulations.
Collapse
Affiliation(s)
- Shahabuddin
- Department of Applied Chemistry, Z. H. College of Engineering & Technology, Aligarh Muslim University Aligarh 202002 India
| | - Uzma
- Division of Organic Synthesis, Department of Chemistry, Aligarh Muslim University Aligarh 202002 India
| | - Mohammad Azam
- Department of Chemistry, College of Science, King Saud University PO 2455 Riyadh 11451 Saudi Arabia
| | - Mehtab Parveen
- Division of Organic Synthesis, Department of Chemistry, Aligarh Muslim University Aligarh 202002 India
| | - Nurul Huda Abd Kadir
- Faculty of Science and Environmental Marine, Universiti Malaysia Terengganu 21030 Terengganu Malaysia
| | - Kim Min
- Department of Safety Engineering, Dongguk University 123 Dongdae-ro Gyeongju-si Gyeongbuk 780714 South Korea
| | - Mahboob Alam
- Department of Safety Engineering, Dongguk University 123 Dongdae-ro Gyeongju-si Gyeongbuk 780714 South Korea
| |
Collapse
|
5
|
Garsi JB, Hocine S, Hensienne R, Moitessier M, Denton H, Major LL, Smith TK, Hanessian S. Revisiting the dipeptidyl carboxypeptidase inhibitor captopril as a source of pan anti-trypanosomatid agents. Bioorg Med Chem Lett 2024; 110:129883. [PMID: 39013490 DOI: 10.1016/j.bmcl.2024.129883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024]
Abstract
The protozoan parasites Trypanosoma brucei, Trypanosoma cruzi and Leishmania spp. are responsible for continued propagation of neglected tropical diseases such as African sleeping sickness, Chagas disease and leishmaniasis respectively. Following a report that captopril targets Leishmania donovani dipeptidyl carboxypeptidase, a series of simple proline amides and captopril analogues were synthesized and found to exhibit 1-2 μM in vitro inhibition and selectivity against Trypanosoma brucei, Trypanosoma cruzi and Leishmania spp. The results were corroborated with computational docking studies. Arguably, the synthetic proline amides represent the structurally simplest examples of in vitro pan antiprotozoal compounds.
Collapse
Affiliation(s)
- Jean-Baptiste Garsi
- Department of Chemistry, Université de Montréal, Station Centre-Ville, C.P. 6128, Montreal, QC H3C 3J7, Canada
| | - Sofiane Hocine
- Department of Chemistry, Université de Montréal, Station Centre-Ville, C.P. 6128, Montreal, QC H3C 3J7, Canada
| | - Raphaël Hensienne
- Department of Chemistry, Université de Montréal, Station Centre-Ville, C.P. 6128, Montreal, QC H3C 3J7, Canada
| | - Matthieu Moitessier
- Department of Chemistry, Université de Montréal, Station Centre-Ville, C.P. 6128, Montreal, QC H3C 3J7, Canada
| | - Helen Denton
- Schools of Biology and Chemistry, Biomedical Sciences Research Complex, University of St. Andrews, St. Andrews, Fife, Scotland KY16 9ST, UK
| | - Louise L Major
- Schools of Biology and Chemistry, Biomedical Sciences Research Complex, University of St. Andrews, St. Andrews, Fife, Scotland KY16 9ST, UK
| | - Terry K Smith
- Schools of Biology and Chemistry, Biomedical Sciences Research Complex, University of St. Andrews, St. Andrews, Fife, Scotland KY16 9ST, UK.
| | - Stephen Hanessian
- Department of Chemistry, Université de Montréal, Station Centre-Ville, C.P. 6128, Montreal, QC H3C 3J7, Canada.
| |
Collapse
|
6
|
Khan H, Hakami MA, Alamri MA, Alotaibi BS, Ullah N, Khan R, Khalid A, Abdalla AN, Wadood A. Identification of Novel Antileishmanial Chemotypes By High-Throughput Virtual and In Vitro Screening. Acta Parasitol 2024; 69:1439-1457. [PMID: 39150581 DOI: 10.1007/s11686-024-00899-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 07/30/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Leishmaniasis is a deadly protozoan parasitic disease and a significant health problem in underdeveloped and developing countries. The global spread of the parasite, coupled with the emergence of drug resistance and severe side effects associated with existing treatments, has necessitated the identification of new and potential drugs. OBJECTIVE This study aimed to identify promising compounds for the treatment of leishmaniasis by targeting two essential enzymes of Leishmania donovani: trypanothione reductase (Try-R) and trypanothione synthetase (Try-S). METHODS High-throughput virtual and in vitro screening of in-house and commercial databases was conducted. A pharmacophore model with seven features was developed and validated using the Guner-Henery method. The pharmacophore-based virtual screening yielded 690 hits, which were further filtered through Lipinski's rule, ADMET analysis, and molecular docking against Try-R and Try-S. Molecular dynamics studies were performed on selected compounds, and in vitro experiments were conducted to evaluate their activity against the promastigote and amastigote forms of L. donovani. RESULTS The virtual screening and subsequent analysis identified 33 promising compounds. Molecular dynamics studies of two compounds (comp-1 and comp-2) demonstrated stable binding interactions with the target enzymes and high affinity. In vitro experiments revealed that 13 compounds exhibited moderate activity against both the promastigote (IC50, 41 µM-76 µM) and the amastigote (IC50, 44 µM-72 µM) forms of L. donovani. Compounds 1 and 2 showed the highest percent inhibition and the lowest IC50 values. CONCLUSION The identified compounds demonstrated significant inhibitory activity against Leishmania donovani and stable interactions with target enzymes. These findings suggest that the compounds could serve as promising leads for developing new treatments for leishmaniasis.
Collapse
Affiliation(s)
- Huma Khan
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra Univesity, Al-Quwayiyah-19257, Riyadh, Saudi Arabia
| | - Mubarak A Alamri
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Bader S Alotaibi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra Univesity, Al-Quwayiyah-19257, Riyadh, Saudi Arabia
| | - Nazif Ullah
- Department of Biotechnology, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Rasool Khan
- Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, 45142, Jazan, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, 21955, Makkah, Saudi Arabia
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan.
| |
Collapse
|
7
|
Singh A, Beg MA, Jamal S, Khan A, Rahman A, Selvapandiyan A, Shafi S, Hoda N. Robust leishmanicidal upshot of some new diphenyl triazine-based molecules. RSC Adv 2024; 14:22587-22597. [PMID: 39021460 PMCID: PMC11253633 DOI: 10.1039/d4ra01904k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
Amongst the neglected tropical diseases, leishmaniasis alone causes 30 000 deaths annually due to the protozoan parasite genus Leishmania. Existing therapies have serious drawbacks in safety, drug resistance, field-adapted application and cost. Therefore, new safer and shorter treatments are an urgent need of the time. Herein, we report the synthesis of fifteen novel diphenyl triazine and diphenyl triazine pyrimidine derivatives and their antileishmanial properties against Leishmania donovani, that causes fatal visceral leishmaniasis. Most of the synthesized analogues exhibited more than 90% inhibition against the promastigote stage of the parasite. Moreover, compounds T4 and T7 showed potent activity against extracellular promastigote (IC50 = 1.074 μM and IC50 = 1.158 μM) as compared to miltefosine (IC50 = 1.477 μM) and is nontoxic towards the host THP-1 macrophage cell line. Interestingly, compound T4 exhibited significant activity against amastigotes (7.186 μM) and induced the macrophages to prevent the survival of the parasite. Our results indicate that T4 represents a new structural lead for this serious and neglected disease.
Collapse
Affiliation(s)
- Anju Singh
- Department of Chemistry, Drug Design and Synthesis Lab., Jamia Millia Islamia Jamia Nagar New Delhi 110025 India +0091-11-26985507 +0091-9910200655
| | - Mirza Adil Beg
- Department of Molecular Medicine, Jamia Hamdard New Delhi 110062 India
| | - Samra Jamal
- Department of Biotechnology, Jamia Hamdard New Delhi 110062 India
| | - Arif Khan
- Department of Chemistry, SCLS, Jamia Hamdard New Delhi 110062 India
| | - Abdur Rahman
- Department of Chemistry, Drug Design and Synthesis Lab., Jamia Millia Islamia Jamia Nagar New Delhi 110025 India +0091-11-26985507 +0091-9910200655
| | | | - Syed Shafi
- Department of Chemistry, SCLS, Jamia Hamdard New Delhi 110062 India
| | - Nasimul Hoda
- Department of Chemistry, Drug Design and Synthesis Lab., Jamia Millia Islamia Jamia Nagar New Delhi 110025 India +0091-11-26985507 +0091-9910200655
| |
Collapse
|
8
|
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
|
9
|
Rivas F, Del Mármol C, Scalese G, Pérez Díaz L, Machado I, Blacque O, Salazar F, Coitiño EL, Benítez D, Medeiros A, Comini M, Gambino D. Multifunctional Organometallic Compounds Active against Infective Trypanosomes: Ru(II) Ferrocenyl Derivatives with Two Different Bioactive Ligands. Inorg Chem 2024; 63:11667-11687. [PMID: 38860314 DOI: 10.1021/acs.inorgchem.4c01125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Human African trypanosomiasis (HAT, sleeping sickness) and American trypanosomiasis (Chagas disease) are endemic zoonotic diseases caused by genomically related trypanosomatid protozoan parasites (Trypanosoma brucei and Trypanosoma cruzi, respectively). Just a few old drugs are available for their treatment, with most of them sharing poor safety, efficacy, and pharmacokinetic profiles. Only fexinidazole has been recently incorporated into the arsenal for the treatment of HAT. In this work, new multifunctional Ru(II) ferrocenyl compounds were rationally designed as potential agents against these pathogens by including in a single molecule 1,1'-bis(diphenylphosphino)ferrocene (dppf) and two bioactive bidentate ligands: pyridine-2-thiolato-1-oxide ligand (mpo) and polypyridyl ligands (NN). Three [Ru(mpo)(dppf)(NN)](PF6) compounds and their derivatives with chloride as a counterion were synthesized and fully characterized in solid state and solution. They showed in vitro activity on bloodstream T. brucei (EC50 = 31-160 nM) and on T. cruzi trypomastigotes (EC50 = 190-410 nM). Compounds showed the lowest EC50 values on T. brucei when compared to the whole set of metal-based compounds previously developed by us. In addition, several of the Ru compounds showed good selectivity toward the parasites, particularly against the highly proliferative bloodstream form of T. brucei. Interaction with DNA and generation of reactive oxygen species (ROS) were ruled out as potential targets and modes of action of the Ru compounds. Biochemical assays and in silico analysis led to the insight that they are able to inhibit the NADH-dependent fumarate reductase from T. cruzi. One representative hit induced a mild oxidation of low molecular weight thiols in T. brucei. The compounds were stable for at least 72 h in two different media and more lipophilic than both bioactive ligands, mpo and NN. An initial assessment of the therapeutic efficacy of one of the most potent and selective candidates, [Ru(mpo)(dppf)(bipy)]Cl, was performed using a murine infection model of acute African trypanosomiasis. This hit compound lacks acute toxicity when applied to animals in the dose/regimen described, but was unable to control parasite proliferation in vivo, probably because of its rapid clearance or low biodistribution in the extracellular fluids. Future studies should investigate the pharmacokinetics of this compound in vivo and involve further research to gain deeper insight into the mechanism of action of the compounds.
Collapse
Affiliation(s)
- Feriannys Rivas
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Carolina Del Mármol
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Gonzalo Scalese
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Leticia Pérez Díaz
- Sección Genómica Funcional, Facultad de Ciencias, Universidad de la República, 11400 Montevideo, Uruguay
| | - Ignacio Machado
- Área Química Analítica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Olivier Blacque
- Department of Chemistry, University of Zurich, CH 8057 Zurich, Switzerland
| | - Fabiana Salazar
- Laboratorio de Química Teórica y Computacional (LQTC), Instituto de Química Biológica, Facultad de Ciencias, and Centro de Investigaciones Biomédicas (CeInBio), Universidad de la República, 11400 Montevideo, Uruguay
| | - E Laura Coitiño
- Laboratorio de Química Teórica y Computacional (LQTC), Instituto de Química Biológica, Facultad de Ciencias, and Centro de Investigaciones Biomédicas (CeInBio), Universidad de la República, 11400 Montevideo, Uruguay
| | - Diego Benítez
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Andrea Medeiros
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, 11800 Montevideo, Uruguay
| | - Marcelo Comini
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Dinorah Gambino
- Área Química Inorgánica, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| |
Collapse
|
10
|
Corfu AI, Santarem N, Luelmo S, Mazza G, Greco A, Altomare A, Ferrario G, Nasta G, Keminer O, Aldini G, Tamborini L, Basilico N, Parapini S, Gul S, Cordeiro-da-Silva A, Conti P, Borsari C. Discovery of 1,3,4-Oxadiazole Derivatives as Broad-Spectrum Antiparasitic Agents. ACS Infect Dis 2024; 10:2222-2238. [PMID: 38717116 DOI: 10.1021/acsinfecdis.4c00181] [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] [Indexed: 06/15/2024]
Abstract
Vector-borne parasitic diseases (VBPDs) pose a significant threat to public health on a global scale. Collectively, Human African Trypanosomiasis (HAT), Leishmaniasis, and Malaria threaten millions of people, particularly in developing countries. Climate change might alter the transmission and spread of VBPDs, leading to a global burden of these diseases. Thus, novel agents are urgently needed to expand therapeutic options and limit the spread of drug-resistant parasites. Herein, we report the development of broad-spectrum antiparasitic agents by screening a known library of antileishmanial and antimalarial compounds toward Trypanosoma brucei (T. brucei) and identifying a 1,3,4-oxadiazole derivative (19) as anti-T. brucei hit with predicted blood-brain barrier permeability. Subsequently, extensive structure-activity-relationship studies around the lipophilic tail of 19 led to a potent antitrypanosomal and antimalarial compound (27), with moderate potency also toward Leishmania infantum (L. infantum) and Leishmania tropica. In addition, we discovered a pan-active antiparasitic molecule (24), showing low-micromolar IC50s toward T. brucei and Leishmania spp. promastigotes and amastigotes, and nanomolar IC50 against Plasmodium falciparum, together with high selectivity for the parasites over mammalian cells (THP-1). Early ADME-toxicity assays were used to assess the safety profile of the compounds. Overall, we characterized 24 and 27, bearing the 1,3,4-oxadiazole privileged scaffold, as broad-spectrum low-toxicity agents for the treatment of VBPDs. An alkyne-substituted chemical probe (30) was synthesized and will be utilized in proteomics experiments aimed at deconvoluting the mechanism of action in the T. brucei parasite.
Collapse
Affiliation(s)
- Alexandra Ioana Corfu
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Nuno Santarem
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sara Luelmo
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Gaia Mazza
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Alessandro Greco
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Alessandra Altomare
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Giulio Ferrario
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Giulia Nasta
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Oliver Keminer
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Lucia Tamborini
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Nicoletta Basilico
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Silvia Parapini
- Department of Biomedical Sciences for Health, University of Milan, Via Pascal 36, 20133 Milan, Italy
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Schnackenburgallee 114, 22525 Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Anabela Cordeiro-da-Silva
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Paola Conti
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Chiara Borsari
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| |
Collapse
|
11
|
Centanni A, Diotallevi A, Buffi G, Olivieri D, Santarém N, Lehtinen A, Yli-Kauhaluoma J, Cordeiro-da-Silva A, Kiuru P, Lucarini S, Galluzzi L. Exploring hydrophilic 2,2-di(indol-3-yl)ethanamine derivatives against Leishmania infantum. PLoS One 2024; 19:e0301901. [PMID: 38870204 PMCID: PMC11175440 DOI: 10.1371/journal.pone.0301901] [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: 01/16/2024] [Accepted: 03/25/2024] [Indexed: 06/15/2024] Open
Abstract
Herein we report the design and the synthesis of a library of new and more hydrophilic bisindole analogues based on our previously identified antileishmanial compound URB1483 that failed the preliminary in vivo test. The novel bisindoles were phenotypically screened for efficacy against Leishmania infantum promastigotes and simultaneously for toxicity on human macrophage-like THP-1 cells. Among the less toxic compounds, eight bisindoles showed IC50 below 10 μM. The most selective compound 1h (selectivity index = 10.1, comparable to miltefosine) and the most potent compound 2c (IC50 = 2.7 μM) were tested for their efficacy on L. infantum intracellular amastigotes. The compounds also demonstrated their efficacy in the in vitro infection model, showing IC50 of 11.1 and 6.8 μM for 1h and 2c, respectively. Moreover, 1h showed a better toxicity profile than the commercial drug miltefosine. For all these reasons, 1h could be a possible new starting point for hydrophilic antileishmanial agents with low cytotoxicity on human macrophage-like cells.
Collapse
Affiliation(s)
- Alessia Centanni
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Aurora Diotallevi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Italy
| | - Gloria Buffi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Italy
| | - Diego Olivieri
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Italy
| | - Nuno Santarém
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto, R. Alfredo Allen, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
| | - Antti Lehtinen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Anabela Cordeiro-da-Silva
- Instituto de Investigação e Inovação em Saúde da Universidade do Porto, R. Alfredo Allen, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
| | - Paula Kiuru
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Simone Lucarini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Italy
| | - Luca Galluzzi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino (PU), Italy
| |
Collapse
|
12
|
Savardashtaki A, Khalili Alashti S, Vafadar A, Sadeghi M, Baneshi M, Hashemi KS, Karami J, Muro A, Manzano-Roman R, Rashidi S. An integrated bioinformatic analysis of microarray datasets to identify biomarkers and miRNA-based regulatory networks in leishmaniasis. Sci Rep 2024; 14:12981. [PMID: 38839916 PMCID: PMC11153516 DOI: 10.1038/s41598-024-63462-5] [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: 11/13/2023] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Micro RNAs (miRNAs, miRs) and relevant networks might exert crucial functions during differential host cell infection by the different Leishmania species. Thus, a bioinformatic analysis of microarray datasets was developed to identify pivotal shared biomarkers and miRNA-based regulatory networks for Leishmaniasis. A transcriptomic analysis by employing a comprehensive set of gene expression profiling microarrays was conducted to identify the key genes and miRNAs relevant for Leishmania spp. infections. Accordingly, the gene expression profiles of healthy human controls were compared with those of individuals infected with Leishmania mexicana, L. major, L. donovani, and L. braziliensis. The enrichment analysis for datasets was conducted by utilizing EnrichR database, and Protein-Protein Interaction (PPI) network to identify the hub genes. The prognostic value of hub genes was assessed by using receiver operating characteristic (ROC) curves. Finally, the miRNAs that interact with the hub genes were identified using miRTarBase, miRWalk, TargetScan, and miRNet. Differentially expressed genes were identified between the groups compared in this study. These genes were significantly enriched in inflammatory responses, cytokine-mediated signaling pathways and granulocyte and neutrophil chemotaxis responses. The identification of hub genes of recruited datasets suggested that TNF, SOCS3, JUN, TNFAIP3, and CXCL9 may serve as potential infection biomarkers and could deserve value as prognostic biomarkers for leishmaniasis. Additionally, inferred data from miRWalk revealed a significant degree of interaction of a number of miRNAs (hsa-miR-8085, hsa-miR-4673, hsa-miR-4743-3p, hsa-miR-892c-3p, hsa-miR-4644, hsa-miR-671-5p, hsa-miR-7106-5p, hsa-miR-4267, hsa-miR-5196-5p, and hsa-miR-4252) with the majority of the hub genes, suggesting such miRNAs play a crucial role afterwards parasite infection. The hub genes and hub miRNAs identified in this study could be potentially suggested as therapeutic targets or biomarkers for the management of leishmaniasis.
Collapse
Affiliation(s)
- Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shayan Khalili Alashti
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboubeh Sadeghi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Baneshi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kimia Sadat Hashemi
- Department of Medical Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Jafar Karami
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran
- Department of Medical Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran
| | - Antonio Muro
- Infectious and Tropical Diseases Group (E-INTRO), Faculty of Pharmacy, Institute of Biomedical Research of Salamanca-Research Center for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), University of Salamanca, 37008, Salamanca, Spain
| | - Raúl Manzano-Roman
- Infectious and Tropical Diseases Group (E-INTRO), Faculty of Pharmacy, Institute of Biomedical Research of Salamanca-Research Center for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), University of Salamanca, 37008, Salamanca, Spain.
| | - Sajad Rashidi
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran.
- Department of Medical Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran.
| |
Collapse
|
13
|
Kumari D, Kour P, Singh CP, Choudhary R, Ali SM, Bhayye S, Bharitkar YP, Singh K. Anhydroparthenin as a dual-target inhibitor against Sterol C-24 methyltransferase and Sterol 14-α demethylase of Leishmania donovani: A comprehensive in vitro and in silico study. Int J Biol Macromol 2024; 269:132034. [PMID: 38702006 DOI: 10.1016/j.ijbiomac.2024.132034] [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: 02/14/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Parthenium hysterophorus plant has a diverse chemical profile and immense bioactive potential. It exhibits excellent pharmacological properties such as anti-cancer, anti-inflammatory, anti-malarial, microbicidal, and anti-trypanosomal. The present study aims to evaluate the anti-leishmanial potential and toxicological safety of anhydroparthenin isolated from P. hysterophorus. Anydroparthenin was extracted from the leaves of P. hysterophorus and characterized through detailed analysis of 1H, 13C NMR, and HRMS. Dye-based in vitro and ex vivo assays confirmed that anhydroparthenin significantly inhibited both promastigote and amastigote forms of the Leishmania donovani parasites. Both the cytotoxicity experiment and hemolytic assay revealed its non-toxic nature and safety index in the range of 10 to 15. Further, various mechanistic assays suggested that anhydroparthenin led to the generation of oxidative stress, intracellular ATP depletion, alterations in morphology and mitochondrial membrane potential, formation of intracellular lipid bodies, and acidic vesicles, ultimately leading to parasite death. As a dual targeting approach, computational studies and sterol quantification assays confirmed that anhydroparthenin inhibits the Sterol C-24 methyl transferase and Sterol 14-α demethylase proteins involved in the ergosterol biosynthesis in Leishmania parasites. These results suggest that anhydroparthenin could be a promising anti-leishmanial molecule and can be developed as a novel therapeutic stratagem against leishmaniasis.
Collapse
Affiliation(s)
- Diksha Kumari
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Parampreet Kour
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Chetan Paul Singh
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Natural Products & Medicinal Chemistry Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Rinku Choudhary
- Department of Bioinformatics, Rajiv Gandhi Institute of I.T. and Biotechnology, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra 411046, India
| | - Syed Mudassir Ali
- Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Sagar Bhayye
- Department of Bioinformatics, Rajiv Gandhi Institute of I.T. and Biotechnology, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra 411046, India
| | - Yogesh P Bharitkar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Natural Products & Medicinal Chemistry Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India.
| | - Kuljit Singh
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
14
|
Worku KM, Asfaw BG, Mamo DN, Haile Y, Tesfa H, Aemero M. Medicinal plants with promising antileishmanial activity in Ethiopia: A systematic review and meta-analysis. Medicine (Baltimore) 2024; 103:e38480. [PMID: 39259058 PMCID: PMC11142835 DOI: 10.1097/md.0000000000038480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 09/12/2024] Open
Abstract
INTRODUCTION Toxicity and resistance to chemotherapy used to treat leishmaniasis are increasing. Research on natural plant compounds has revealed their antileishmanial effects on certain Leishmania organisms. This review aimed to estimate the pooled IC50 values of medicinal plants with promising antileishmanial activity in Ethiopia. METHODS A systematic literature search was conducted using Science Direct, PubMed, Cochrane Library, and Google Scholar to locate potential studies. Studies published in peer-reviewed journals and gray literature in university repositories before April 1, 2022, which included a full-length study reporting the half-maximal inhibitory concentration (IC50) of Ethiopian medicinal plants that were written in English were included. Conference proceedings, review articles, letters to the editor, and correspondence were excluded. The quality of the included studies was assessed using the GIVIMP critical appraisal tools. Heterogeneity between studies was verified using Cochrane Q test statistics and I2 test statistics, and the effects were checked using Egger statistical test at a level of significance. A random-effects model was used to estimate the pooled IC50 of the medicinal plants. RESULTS Six articles that were conducted in Ethiopia that fulfilled the inclusion criteria, with a total of 62 in vitro experiments, were reviewed. The aggregated mean IC50 for medicinal plants in Ethiopia was 16.80 (95% CI: 12.44, 21.16) and 13.81 (95% CI: 13.12, 14.50) µg/mL for antipromastigote and antiamastigote activity, respectively. Aqueous was the significant preparation with IC50 of 0.53 (0.34, 0.73) µg/mL against promastigote and 0.98 (0.20, 1.76) µg/mL against the amastigote stage. DISCUSSION This review indicated that the pooled mean of IC50 for Ethiopian medicinal plants against promastigotes and amastigotes was relatively low and showed better efficacy. This strongly suggests the need to focus on antipromastigote and antiamastigote medicinal plants in Ethiopia for the development of antileishmanial drugs. It is necessary to identify their active components, and their potential toxic effects can lead to the production of well-tolerated and safe drugs for leishmaniasis. The high heterogeneity is the limitation of this study. REGISTRATION The review has been registered at Prospero with identification number CRD42022343543.
Collapse
Affiliation(s)
- Kassahun Misgana Worku
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, Ethiopia
| | - Birhanu Genanew Asfaw
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, Ethiopia
| | - Daniel Niguse Mamo
- Department of Health Informatics, School of Public Health, College of Medicine and Health Science, Arba Minch University, Arba Minch, Ethiopia
| | - Yosef Haile
- Department of Public Health, School of Public Health, College of Medicine and Health Science, Arba Minch University, Arba Minch, Ethiopia
| | - Habtie Tesfa
- Department of Medical Parasitology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Mulugeta Aemero
- Department of Medical Parasitology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
15
|
Kancharla P, Ortiz D, Fargo CM, Zhang X, Li Y, Sanchez M, Kumar A, Yeluguri M, Dodean RA, Caridha D, Madejczyk MS, Martin M, Jin X, Blount C, Chetree R, Pannone K, Dinh HT, DeLuca J, Evans M, Nadeau R, Vuong C, Leed S, Dennis WE, Roncal N, Pybus BS, Lee PJ, Roth A, Reynolds KA, Kelly JX, Landfear SM. Discovery and Optimization of Tambjamines as a Novel Class of Antileishmanial Agents. J Med Chem 2024; 67:8323-8345. [PMID: 38722757 PMCID: PMC11163866 DOI: 10.1021/acs.jmedchem.4c00517] [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] [Indexed: 05/24/2024]
Abstract
Leishmaniasis is a neglected tropical disease that is estimated to afflict over 12 million people. Current drugs for leishmaniasis suffer from serious deficiencies, including toxicity, high cost, modest efficacy, primarily parenteral delivery, and emergence of widespread resistance. We have discovered and developed a natural product-inspired tambjamine chemotype, known to be effective against Plasmodium spp, as a novel class of antileishmanial agents. Herein, we report in vitro and in vivo antileishmanial activities, detailed structure-activity relationships, and metabolic/pharmacokinetic profiles of a large library of tambjamines. A number of tambjamines exhibited excellent potency against both Leishmania mexicana and Leishmania donovani parasites with good safety and metabolic profiles. Notably, tambjamine 110 offered excellent potency and provided partial protection to leishmania-infected mice at 40 and/or 60 mg/kg/10 days of oral treatment. This study presents the first account of antileishmanial activity in the tambjamine family and paves the way for the generation of new oral antileishmanial drugs.
Collapse
Affiliation(s)
- Papireddy Kancharla
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Diana Ortiz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, 97239, United States
| | - Corinne M. Fargo
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, 97239, United States
| | - Xiaowei Zhang
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Yuexin Li
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Marco Sanchez
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, 97239, United States
| | - Amrendra Kumar
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Monish Yeluguri
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Rozalia A. Dodean
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Diana Caridha
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Michael S. Madejczyk
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Monica Martin
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Xiannu Jin
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Cameron Blount
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Ravi Chetree
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Kristina Pannone
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Hieu T. Dinh
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Jesse DeLuca
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Martin Evans
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Robert Nadeau
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Chau Vuong
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Susan Leed
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - William E. Dennis
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Norma Roncal
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Brandon S. Pybus
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Patricia J. Lee
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Alison Roth
- Experimental Therapeutics Branch, CIDR, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Kevin A. Reynolds
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
| | - Jane X. Kelly
- Department of Chemistry, Portland State University, Portland, Oregon 97201, United States
- Department of Veterans Affairs Medical Center, Portland, Oregon 97239, United States
| | - Scott M. Landfear
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, 97239, United States
| |
Collapse
|
16
|
Camargo PG, Dos Santos CR, Girão Albuquerque M, Rangel Rodrigues C, Lima CHDS. Py-CoMFA, docking, and molecular dynamics simulations of Leishmania (L.) amazonensis arginase inhibitors. Sci Rep 2024; 14:11575. [PMID: 38773273 PMCID: PMC11109165 DOI: 10.1038/s41598-024-62520-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/17/2024] [Indexed: 05/23/2024] Open
Abstract
Leishmaniasis is a disease caused by a protozoan of the genus Leishmania, affecting millions of people, mainly in tropical countries, due to poor social conditions and low economic development. First-line chemotherapeutic agents involve highly toxic pentavalent antimonials, while treatment failure is mainly due to the emergence of drug-resistant strains. Leishmania arginase (ARG) enzyme is vital in pathogenicity and contributes to a higher infection rate, thus representing a potential drug target. This study helps in designing ARG inhibitors for the treatment of leishmaniasis. Py-CoMFA (3D-QSAR) models were constructed using 34 inhibitors from different chemical classes against ARG from L. (L.) amazonensis (LaARG). The 3D-QSAR predictions showed an excellent correlation between experimental and calculated pIC50 values. The molecular docking study identified the favorable hydrophobicity contribution of phenyl and cyclohexyl groups as substituents in the enzyme allosteric site. Molecular dynamics simulations of selected protein-ligand complexes were conducted to understand derivatives' interaction modes and affinity in both active and allosteric sites. Two cinnamide compounds, 7g and 7k, were identified, with similar structures to the reference 4h allosteric site inhibitor. These compounds can guide the development of more effective arginase inhibitors as potential antileishmanial drugs.
Collapse
Affiliation(s)
- Priscila Goes Camargo
- Faculdade de Farmácia, Departamento de Fármacos e Medicamentos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carine Ribeiro Dos Santos
- Laboratório de Modelagem Molecular (LabMMol), Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Magaly Girão Albuquerque
- Laboratório de Modelagem Molecular (LabMMol), Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carlos Rangel Rodrigues
- Faculdade de Farmácia, Departamento de Fármacos e Medicamentos, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Camilo Henrique da Silva Lima
- Laboratório de Modelagem Molecular (LabMMol), Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
17
|
Marín M, López M, Gallego-Yerga L, Álvarez R, Peláez R. Experimental structure based drug design (SBDD) applications for anti-leishmanial drugs: A paradigm shift? Med Res Rev 2024; 44:1055-1120. [PMID: 38142308 DOI: 10.1002/med.22005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 12/25/2023]
Abstract
Leishmaniasis is a group of neglected tropical diseases caused by at least 20 species of Leishmania protozoa, which are spread by the bite of infected sandflies. There are three main forms of the disease: cutaneous leishmaniasis (CL, the most common), visceral leishmaniasis (VL, also known as kala-azar, the most serious), and mucocutaneous leishmaniasis. One billion people live in areas endemic to leishmaniasis, with an annual estimation of 30,000 new cases of VL and more than 1 million of CL. New treatments for leishmaniasis are an urgent need, as the existing ones are inefficient, toxic, and/or expensive. We have revised the experimental structure-based drug design (SBDD) efforts applied to the discovery of new drugs against leishmaniasis. We have grouped the explored targets according to the metabolic pathways they belong to, and the key achieved advances are highlighted and evaluated. In most cases, SBDD studies follow high-throughput screening campaigns and are secondary to pharmacokinetic optimization, due to the majoritarian belief that there are few validated targets for SBDD in leishmaniasis. However, some SBDD strategies have significantly contributed to new drug candidates against leishmaniasis and a bigger number holds promise for future development.
Collapse
Affiliation(s)
- Miguel Marín
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Marta López
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Laura Gallego-Yerga
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| |
Collapse
|
18
|
Khairnar P, Saathoff JM, Cook DW, Hochstetler SR, Pandya U, Robinson SJ, Satam V, Donsbach KO, Gupton BF, Jin LM, Shanahan CS. Practical Synthesis of 6-Amino-1-hydroxy-2,1-benzoxaborolane: A Key Intermediate of DNDI-6148. Org Process Res Dev 2024; 28:1213-1223. [PMID: 38660377 PMCID: PMC11036395 DOI: 10.1021/acs.oprd.4c00031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/03/2024] [Accepted: 03/11/2024] [Indexed: 04/26/2024]
Abstract
Visceral leishmaniasis (VL), a parasitic, poverty-linked, neglected disease, is endemic across multiple regions of the world and fatal if untreated. There is an urgent need for a better and more affordable treatment for VL. DNDI-6148 is a promising drug candidate being evaluated for the treatment of VL; however, the current process for producing the key intermediate of DNDI-6148, 6-amino-1-hydroxy-2,1-benzoxaborolane, is expensive and difficult to scale up. Herein, we describe two practical approaches to synthesizing 6-amino-1-hydroxy-2,1-benzoxaborolane from inexpensive and readily available raw materials. Starting with 4-tolunitrile, the first approach is a five-step sequence involving a Hofmann rearrangement, resulting in an overall yield of 40%. The second approach utilizes 2-methyl-5-nitroaniline as the starting material and features borylation of aniline and continuous flow hydrogenation as the key steps, with an overall yield of 46%. Both routes bypass the nitration of 1-hydroxy-2,1-benzoxaborolane, which is challenging and expensive to scale. In particular, the second approach is more practical and scalable because of the mild operating conditions and facile isolation process.
Collapse
Affiliation(s)
- Pankaj
V. Khairnar
- Medicines
for All Institute, Virginia Commonwealth
University, Richmond, Virginia 23284-3068, United States
| | - John M. Saathoff
- Medicines
for All Institute, Virginia Commonwealth
University, Richmond, Virginia 23284-3068, United States
| | - Daniel W. Cook
- Medicines
for All Institute, Virginia Commonwealth
University, Richmond, Virginia 23284-3068, United States
| | - Samuel R. Hochstetler
- Medicines
for All Institute, Virginia Commonwealth
University, Richmond, Virginia 23284-3068, United States
| | - Urvish Pandya
- Drugs
for Neglected Diseases initiative, 15 Chemin Camille-Vidart, 1202 Geneva, Switzerland
| | - Stephen J. Robinson
- Drugs
for Neglected Diseases initiative, 15 Chemin Camille-Vidart, 1202 Geneva, Switzerland
| | - Vijay Satam
- Drugs
for Neglected Diseases initiative, 15 Chemin Camille-Vidart, 1202 Geneva, Switzerland
| | - Kai O. Donsbach
- Medicines
for All Institute, Virginia Commonwealth
University, Richmond, Virginia 23284-3068, United States
| | - B. Frank Gupton
- Medicines
for All Institute, Virginia Commonwealth
University, Richmond, Virginia 23284-3068, United States
| | - Li-Mei Jin
- Medicines
for All Institute, Virginia Commonwealth
University, Richmond, Virginia 23284-3068, United States
| | - Charles S. Shanahan
- Medicines
for All Institute, Virginia Commonwealth
University, Richmond, Virginia 23284-3068, United States
| |
Collapse
|
19
|
Essid R, Kefi S, Damergi B, Abid G, Fares N, Jallouli S, Abid I, Hussein D, Tabbene O, Limam F. Promising Antileishmanial Activity of Micromeria nervosa Essential Oil: In Vitro and In Silico Studies. Molecules 2024; 29:1876. [PMID: 38675696 PMCID: PMC11055018 DOI: 10.3390/molecules29081876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/17/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
The present study aimed to evaluate the leishmanicidal potential of the essential oil (EO) of Micromeria (M.) nervosa and to investigate its molecular mechanism of action by qPCR. Furthermore, in silicointeraction study of the major M. nervosa EO compounds with the enzyme cytochrome P450 sterol 14α-demethylase (CYP51) was also performed. M. nervosa EO was analyzed by gas chromatography-mass spectrometry (GC-MS). Results showed that α-pinene (26.44%), t-cadinol (26.27%), caryophyllene Oxide (7.73 ± 1.04%), and α-Cadinene (3.79 ± 0.12%) are the major compounds of M. nervosa EO. However, limited antioxidant activity was observed, as this EO was ineffective in neutralizing DPPH free radicals and in inhibiting β-carotene bleaching. Interestingly, it displayed effective leishmanicidal potential against promastigote (IC50 of 6.79 and 5.25 μg/mL) and amastigote (IC50 of 8.04 and 7.32 μg/mL) forms of leishmania (L.) infantum and L. major, respectively. Molecular mechanism investigation showed that M. nervosa EO displayed potent inhibition on the thiol regulatory pathway. Furthermore, a docking study of the main components of the EO with cytochrome P450 sterol 14α-demethylase (CYP51) enzyme revealed that t-cadinol exhibited the best binding energy values (-7.5 kcal/mol), followed by α-cadinene (-7.3 kcal/mol) and caryophyllene oxide (-7 kcal/mol). These values were notably higher than that of the conventional drug fluconazole showing weaker binding energy (-6.9 kcal/mol). These results suggest that M. nervosa EO could serve as a potent and promising candidate for the development of alternative antileishmanial agent in the treatment of leishmaniasis.
Collapse
Affiliation(s)
- Rym Essid
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP 901, Hammam-Lif 2050, Tunisia; (S.K.); (N.F.)
- University of Tunis-El Manar, Campus Universitaire Farhat Hached, BP-94 Rommana, Tunis 1068, Tunisia
| | - Sarra Kefi
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP 901, Hammam-Lif 2050, Tunisia; (S.K.); (N.F.)
- University of Tunis-El Manar, Campus Universitaire Farhat Hached, BP-94 Rommana, Tunis 1068, Tunisia
| | - Bilel Damergi
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP 901, Hammam-Lif 2050, Tunisia; (S.K.); (N.F.)
- University of Tunis-El Manar, Campus Universitaire Farhat Hached, BP-94 Rommana, Tunis 1068, Tunisia
| | - Ghassen Abid
- Laboratory of Legumes and Sustainable Agro-Systems, Centre of Biotechnology of Borj Cedria, Hammam-Lif 2050, Tunisia
| | - Nadia Fares
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP 901, Hammam-Lif 2050, Tunisia; (S.K.); (N.F.)
| | - Selim Jallouli
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP 901, Hammam-Lif 2050, Tunisia; (S.K.); (N.F.)
| | - Islem Abid
- Center of Excellence in Biotechnology Research, College of Applied Medical Sciences, King Saud University, Riyadh 11495, Saudi Arabia
| | - Dina Hussein
- Department of Chemistry, College of Sciences and Health, Cleveland State University, Cleveland, OH 44115, USA;
| | - Olfa Tabbene
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP 901, Hammam-Lif 2050, Tunisia; (S.K.); (N.F.)
| | - Ferid Limam
- Laboratory of Bioactive Substances, Biotechnology Center of Borj Cedria, BP 901, Hammam-Lif 2050, Tunisia; (S.K.); (N.F.)
| |
Collapse
|
20
|
Lapierre TJWJD, Farago DN, de Moura Lodi Cruz MGF, de Melo Resende D, de Oliveira ACR, Dos Santos BRM, de Oliveira Souza F, Michelan-Duarte S, Chelucci RC, Andricopulo AD, Ferreira LLG, Pilau EJ, Murta SMF, de Oliveira Rezende Júnior C. Evaluation and discovery of novel benzothiazole derivatives as promising hits against Leishmania infantum. Chem Biol Drug Des 2024; 103:e14525. [PMID: 38627214 DOI: 10.1111/cbdd.14525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/23/2024] [Accepted: 04/05/2024] [Indexed: 04/19/2024]
Abstract
An early exploration of the benzothiazole class against two kinetoplastid parasites, Leishmania infantum and Trypanosoma cruzi, has been performed after the identification of a benzothiazole derivative as a suitable antileishmanial initial hit. The first series of derivatives focused on the acyl fragment of its class, evaluating diverse linear and cyclic, alkyl and aromatic substituents, and identified two other potent compounds, the phenyl and cyclohexyl derivatives. Subsequently, new compounds were designed to assess the impact of the presence of diverse substituents on the benzothiazole ring or the replacement of the endocyclic sulfur by other heteroatoms. All compounds showed relatively low cytotoxicity, resulting in decent selectivity indexes for the most active compounds. Ultimately, the in vitro ADME properties of these compounds were assessed, revealing a satisfying water solubility, gastrointestinal permeability, despite their low metabolic stability and high lipophilicity. Consequently, compounds 5 and 6 were identified as promising hits for further hit-to-lead exploration within this benzothiazole class against L. infantum, thus providing promising starting points for the development of antileishmanial candidates.
Collapse
Affiliation(s)
| | - Danilo Nascimento Farago
- Laboratório de Síntese de Candidatos a Fármacos, Instituto de Química, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | | | - Daniela de Melo Resende
- Grupo de Genômica Funcional de Parasitos, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ Minas), Belo Horizonte, Minas Gerais, Brazil
| | - Adriane Cristina Rosa de Oliveira
- Laboratório de Síntese de Candidatos a Fármacos, Instituto de Química, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | - Brenda Rosa Macedo Dos Santos
- Laboratório de Síntese de Candidatos a Fármacos, Instituto de Química, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| | - Felipe de Oliveira Souza
- Laboratório de Biomoléculas e Espectrometria de Massas (LaBioMass), Universidade Estadual de Maringá (UEM), Maringá, Paraná, Brazil
| | - Simone Michelan-Duarte
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos, São Paulo, Brazil
| | - Rafael C Chelucci
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos, São Paulo, Brazil
| | - Adriano D Andricopulo
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos, São Paulo, Brazil
| | - Leonardo L G Ferreira
- Laboratório de Química Medicinal e Computacional (LQMC), Instituto de Física de São Carlos (IFSC), Universidade de São Paulo (USP), São Carlos, São Paulo, Brazil
| | - Eduardo Jorge Pilau
- Laboratório de Biomoléculas e Espectrometria de Massas (LaBioMass), Universidade Estadual de Maringá (UEM), Maringá, Paraná, Brazil
| | - Silvane Maria Fonseca Murta
- Grupo de Genômica Funcional de Parasitos, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ Minas), Belo Horizonte, Minas Gerais, Brazil
| | - Celso de Oliveira Rezende Júnior
- Laboratório de Síntese de Candidatos a Fármacos, Instituto de Química, Universidade Federal de Uberlândia (UFU), Uberlândia, Minas Gerais, Brazil
| |
Collapse
|
21
|
Bharadava K, Upadhyay TK, Kaushal RS, Ahmad I, Alraey Y, Siddiqui S, Saeed M. Genomic Insight of Leishmania Parasite: In-Depth Review of Drug Resistance Mechanisms and Genetic Mutations. ACS OMEGA 2024; 9:12500-12514. [PMID: 38524425 PMCID: PMC10955595 DOI: 10.1021/acsomega.3c09400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 03/26/2024]
Abstract
Leishmaniasis, which is caused by a parasitic protozoan of the genus Leishmania, is still a major threat to global health, impacting millions of individuals worldwide in endemic areas. Chemotherapy has been the principal method for managing leishmaniasis; nevertheless, the evolution of drug resistance offers a significant obstacle to therapeutic success. Drug-resistant behavior in these parasites is a complex phenomenon including both innate and acquired mechanisms. Resistance is frequently related to changes in drug transportation, drug target alterations, and enhanced efflux of the drug from the pathogen. This review has revealed specific genetic mutations in Leishmania parasites that are associated with resistance to commonly used antileishmanial drugs such as pentavalent antimonials, miltefosine, amphotericin B, and paromomycin, resulting in changes in gene expression along with the functioning of various proteins involved in drug uptake, metabolism, and efflux. Understanding the genetic changes linked to drug resistance in Leishmania parasites is essential for creating approaches for tackling and avoiding the spread of drug-resistant variants. Based on which specific treatments focus on mutations and pathways could potentially improve treatment efficacy and help long-term leishmaniasis control. More study is needed to uncover the complete range of genetic changes generating medication resistance and to develop new therapies based on available information.
Collapse
Affiliation(s)
- Krupanshi Bharadava
- Biophysics
& Structural Biology, Research & Development Cell, Parul University, Vadodara, Gujarat 391760, India
| | - Tarun Kumar Upadhyay
- Department
of Life Sciences, Parul Institute of Applied Sciences & Research
and Development Cell, Parul University, Vadodara, Gujarat 391760, India
| | - Radhey Shyam Kaushal
- Biophysics
& Structural Biology, Research & Development Cell, Parul University, Vadodara, Gujarat 391760, India
- Department
of Life Sciences, Parul Institute of Applied Sciences & Research
and Development Cell, Parul University, Vadodara, Gujarat 391760, India
| | - Irfan Ahmad
- Department
of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Yasser Alraey
- Department
of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Samra Siddiqui
- Department
of Health Service Management, College of Public Health and Health
Informatics, University of Hail, Hail 55476, Saudi Arabia
| | - Mohd Saeed
- Department
of Biology, College of Science, University
of Hail, Hail 55476, Saudi Arabia
| |
Collapse
|
22
|
Rao K, Abdullah M, Ahmed U, Wehelie HI, Shah MR, Siddiqui R, Khan NA, Alawfi BS, Anwar A. Self-assembled micelles loaded with itraconazole as anti-Acanthamoeba nano-formulation. Arch Microbiol 2024; 206:134. [PMID: 38433145 DOI: 10.1007/s00203-024-03854-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/13/2024] [Accepted: 01/17/2024] [Indexed: 03/05/2024]
Abstract
Acanthamoeba castellanii are opportunistic pathogens known to cause infection of the central nervous system termed: granulomatous amoebic encephalitis, that mostly effects immunocompromised individuals, and a sight threatening keratitis, known as Acanthamoeba keratitis, which mostly affects contact lens wearers. The current treatment available is problematic, and is toxic. Herein, an amphiphilic star polymer with AB2 miktoarms [A = hydrophobic poly(ℇ-Caprolacton) and B = hydrophilic poly (ethylene glycol)] was synthesized by ring opening polymerization and CuI catalyzed azide-alkyne cycloaddition. Characterization by 1H and 13C NMR spectroscopy, size-exclusion chromatography and fluorescence spectroscopy was accomplished. The hydrophobic drug itraconazole (ITZ) was incorporated in self-assembled micellar structure of AB2 miktoarms through co-solvent evaporation. The properties of ITZ loaded (ITZ-PCL-PEG2) and blank micelles (PCL-PEG2) were investigated through zeta sizer, scanning electron microscopy and Fourier-transform infrared spectroscopy. Itraconazole alone (ITZ), polymer (DPB-PCL), empty polymeric micelles (PCL-PEG2) alone, and itraconazole loaded in polymeric micelles (ITZ-PCL-PEG2) were tested for anti-amoebic potential against Acanthamoeba, and the cytotoxicity on human cells were determined. The polymer was able to self-assemble in aqueous conditions and exhibited low value for critical micelle concentration (CMC) 0.05-0.06 µg/mL. The maximum entrapment efficiency of ITZ was 68%. Of note, ITZ, DPB, PCL-PEG2 and ITZ-PCL-PEG2 inhibited amoebae trophozoites by 37.34%, 36.30%, 35.77%, and 68.24%, respectively, as compared to controls. Moreover, ITZ-PCL-PEG2 revealed limited cytotoxicity against human keratinocyte cells. These results are indicative that ITZ-PCL-PEG2 micelle show significantly better anti-amoebic effects as compared to ITZ alone and thus should be investigated further in vivo to determine its clinical potential.
Collapse
Affiliation(s)
- Komal Rao
- International Center for Chemical and Biological Sciences, HEJ Research Institute of Chemistry, Karachi University, Karachi, 75270, Pakistan
| | - Muhammad Abdullah
- International Center for Chemical and Biological Sciences, HEJ Research Institute of Chemistry, Karachi University, Karachi, 75270, Pakistan
| | - Usman Ahmed
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Hashi Isse Wehelie
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Muhammad Raza Shah
- International Center for Chemical and Biological Sciences, HEJ Research Institute of Chemistry, Karachi University, Karachi, 75270, Pakistan
| | - Ruqaiyyah Siddiqui
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University Edinburgh, Edinburgh, EH14 4AS, UK
- Microbiota Research Center, Istinye University, 34010, Istanbul, Turkey
| | - Naveed A Khan
- Microbiota Research Center, Istinye University, 34010, Istanbul, Turkey.
| | - Bader S Alawfi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, 42353, Madinah, Saudi Arabia
| | - Ayaz Anwar
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
23
|
Matos ÂP, Saldanha-Corrêa FMP, Gomes RDS, Hurtado GR. Exploring microalgal and cyanobacterial metabolites with antiprotozoal activity against Leishmania and Trypanosoma parasites. Acta Trop 2024; 251:107116. [PMID: 38159713 DOI: 10.1016/j.actatropica.2023.107116] [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: 10/25/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Neglected tropical diseases (NTD) like Leishmaniasis and trypanosomiasis affect millions of people annually, while currently used antiprotozoal drugs have serious side effects. Drug research based on natural products has shown that microalgae and cyanobacteria are a promising platform of biochemically active compounds with antiprotozoal activity. These unicellular photosynthetic organisms are rich in polyunsaturated fatty acids, pigments including phycocyanin, chlorophylls and carotenoids, polyphenols, bioactive peptides, terpenes, alkaloids, which have proven antioxidant, antimicrobial, antiviral, antiplasmodial and antiprotozoal properties. This review provides up-to-date information regarding ongoing studies on substances synthesized by microalgae and cyanobacteria with notable activity against Leishmania spp., Trypanosoma cruzi, and Trypanosoma brucei, the causative agents of Leishmaniasis, Chagas disease, and human African trypanosomiasis, respectively. Extracts of several freshwater or marine microalgae have been tested on different strains of Leishmania and Trypanosoma parasites. For instance, ethanolic extract of Chlamydomonas reinhardtii and Tetraselmis suecica have biological activity against T. cruzi, due to their high content of carotenoids, chlorophylls, phenolic compounds and flavonoids that are associated with trypanocidal activity. Halophilic Dunaliella salina showed moderate antileishmanial activity that may be attributed to the high β-carotene content in this microalga. Peptides such as almiramides, dragonamides, and herbamide that are biosynthesized by marine cyanobacteria Lyngbya majuscula were found to have increased activity in micromolar scale IC50 against L. donovani, T. Cruzi, and T. brucei parasites. The cyanobacterial peptides symplocamide and venturamide isolated from Symploca and Oscillatoria species, respectively, and the alkaloid nostocarbonile isolated from Nostoc have shown promising antiprotozoal properties and are being explored for pharmaceutical and medicinal purposes. The discovery of new molecules from microalgae and cyanobacteria with therapeutic potential against Leishmaniasis and trypanosomiasis may address an urgent medical need: effective and safe treatments of NTDs.
Collapse
Affiliation(s)
- Ângelo Paggi Matos
- Institute for Advanced Studies of Ocean, São Paulo State University (UNESP), Rodovia Presidente Dutra Km 138, Eugênio de Melo, São José dos Campos 12247-004, Brazil.
| | | | - Roberto da Silva Gomes
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Gabriela Ramos Hurtado
- Institute for Advanced Studies of Ocean, São Paulo State University (UNESP), Rodovia Presidente Dutra Km 138, Eugênio de Melo, São José dos Campos 12247-004, Brazil; Institute of Science and Technology, São Paulo State University (UNESP), Rodovia Presidente Dutra Km 138, Eugênio de Melo, São José dos Campos 12247-004, Brazil.
| |
Collapse
|
24
|
Berhe H, Kumar Cinthakunta Sridhar M, Zerihun M, Qvit N. The Potential Use of Peptides in the Fight against Chagas Disease and Leishmaniasis. Pharmaceutics 2024; 16:227. [PMID: 38399281 PMCID: PMC10892537 DOI: 10.3390/pharmaceutics16020227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/28/2023] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Chagas disease and leishmaniasis are both neglected tropical diseases that affect millions of people around the world. Leishmaniasis is currently the second most widespread vector-borne parasitic disease after malaria. The World Health Organization records approximately 0.7-1 million newly diagnosed leishmaniasis cases each year, resulting in approximately 20,000-30,000 deaths. Also, 25 million people worldwide are at risk of Chagas disease and an estimated 6 million people are infected with Trypanosoma cruzi. Pentavalent antimonials, amphotericin B, miltefosine, paromomycin, and pentamidine are currently used to treat leishmaniasis. Also, nifurtimox and benznidazole are two drugs currently used to treat Chagas disease. These drugs are associated with toxicity problems such as nephrotoxicity and cardiotoxicity, in addition to resistance problems. As a result, the discovery of novel therapeutic agents has emerged as a top priority and a promising alternative. Overall, there is a need for new and effective treatments for Chagas disease and leishmaniasis, as the current drugs have significant limitations. Peptide-based drugs are attractive due to their high selectiveness, effectiveness, low toxicity, and ease of production. This paper reviews the potential use of peptides in the treatment of Chagas disease and leishmaniasis. Several studies have demonstrated that peptides are effective against Chagas disease and leishmaniasis, suggesting their use in drug therapy for these diseases. Overall, peptides have the potential to be effective therapeutic agents against Chagas disease and leishmaniasis, but more research is needed to fully investigate their potential.
Collapse
Affiliation(s)
| | | | | | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311502, Israel; (H.B.); (M.K.C.S.); (M.Z.)
| |
Collapse
|
25
|
Pal R, Teli G, Akhtar MJ, Matada GSP. Synthetic product-based approach toward potential antileishmanial drug development. Eur J Med Chem 2024; 263:115927. [PMID: 37976706 DOI: 10.1016/j.ejmech.2023.115927] [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: 08/19/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
Leishmaniasis is a parasitic disease and is categorized as a tropically neglected disease (NTD) with no effective vaccines available. The available chemotherapeutics against leishmaniasis are associated with an increase in the incidence of toxicity and drug resistance. Consequently, targeting metabolic pathways and enzymes of parasites which differs from the mammalian host can be exploited to treat and overcome the resistance. The classical methods of identifying the structural fragments and the moieties responsible for the biological activities from the standard compounds and their modification are options for developing more effective novel compounds. Significant progress has been made in refining the development of potent non-toxic molecules and addressing the limitations of the current treatment available. Several examples of synthetic product-based approach utilizing their core heterocyclic rings including furan, pyrrole, thiazole, imidazole, pyrazole, triazole, quinazoline, quinoline, pyrimidine, coumarin, indole, acridine, oxadiazole, purine, chalcone, carboline, phenanthrene and metal containing derivatives and their structure-activity relationships are discussed in this review. It also analyses the groups/fragments interacting with the host cell receptors and will support the medicinal chemists with novel antileishmanial agents.
Collapse
Affiliation(s)
- Rohit Pal
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - Ghanshyam Teli
- School of Pharmacy, Sangam University, Atoon, Bhilwara, 311001, Rajasthan, India
| | - Md Jawaid Akhtar
- Department of Pharmaceutical Chemistry, National University of Science and Technology, PO 620, PC 130, Azaiba Bousher, Muscat, Sultanate of Oman
| | - Gurubasavaraja Swamy Purawarga Matada
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| |
Collapse
|
26
|
Alsharedeh RH, Rezigue M, Bashatwah RM, Amawi H, Aljabali AAA, Obeid MA, Tambuwala MM. Nanomaterials as a Potential Target for Infectious Parasitic Agents. Curr Drug Deliv 2024; 21:828-851. [PMID: 36815647 DOI: 10.2174/1567201820666230223085403] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/29/2022] [Accepted: 11/16/2022] [Indexed: 02/24/2023]
Abstract
Despite the technological advancement in the era of personalized medicine and therapeutics development, infectious parasitic causative agents remain one of the most challenging areas of research and development. The disadvantages of conventional parasitic prevention and control are the emergence of multiple drug resistance as well as the non-specific targeting of intracellular parasites, which results in high dose concentration needs and subsequently intolerable cytotoxicity. Nanotechnology has attracted extensive interest to reduce medication therapy adverse effects including poor bioavailability and drug selectivity. Numerous nanomaterials-based delivery systems have previously been shown in animal models to be effective in the treatment of various parasitic infections. This review discusses a variety of nanomaterials-based antiparasitic procedures and techniques as well as the processes that allow them to be targeted to different parasitic infections. This review focuses on the key prerequisites for creating novel nanotechnology-based carriers as a potential option in parasite management, specifically in the context of human-related pathogenic parasitic agents.
Collapse
Affiliation(s)
- Rawan H Alsharedeh
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, 21163, Jordan
| | - Meriem Rezigue
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, 21163, Jordan
| | - Rasha M Bashatwah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, 21163, Jordan
| | - Haneen Amawi
- Department of Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, 21163, Jordan
| | - Mohammad A Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, 21163, Jordan
| | - Murtaza M Tambuwala
- Lincoln Medical School, Brayford Pool Campus, University of Lincoln, Lincoln LN6 7TS, United Kingdom
| |
Collapse
|
27
|
Barrera-Téllez FJ, Prieto-Martínez FD, Hernández-Campos A, Martínez-Mayorga K, Castillo-Bocanegra R. In Silico Exploration of the Trypanothione Reductase (TryR) of L. mexicana. Int J Mol Sci 2023; 24:16046. [PMID: 38003236 PMCID: PMC10671491 DOI: 10.3390/ijms242216046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/23/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Human leishmaniasis is a neglected tropical disease which affects nearly 1.5 million people every year, with Mexico being an important endemic region. One of the major defense mechanisms of these parasites is based in the polyamine metabolic pathway, as it provides the necessary compounds for its survival. Among the enzymes in this route, trypanothione reductase (TryR), an oxidoreductase enzyme, is crucial for the Leishmania genus' survival against oxidative stress. Thus, it poses as an attractive drug target, yet due to the size and features of its catalytic pocket, modeling techniques such as molecular docking focusing on that region is not convenient. Herein, we present a computational study using several structure-based approaches to assess the druggability of TryR from L. mexicana, the predominant Leishmania species in Mexico, beyond its catalytic site. Using this consensus methodology, three relevant pockets were found, of which the one we call σ-site promises to be the most favorable one. These findings may help the design of new drugs of trypanothione-related diseases.
Collapse
Affiliation(s)
- Francisco J. Barrera-Téllez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Fernando D. Prieto-Martínez
- Instituto de Química, Unidad Mérida, Universidad Nacional Autónoma de México, Carretera Mérida-Tetiz, Km. 4.5, Ucú 97357, Mexico
| | - Alicia Hernández-Campos
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Karina Martínez-Mayorga
- Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Unidad Mérida, Universidad Nacional Autónoma de México, Sierra Papacal, Mérida 97302, Mexico
| | - Rafael Castillo-Bocanegra
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
Collapse
|
28
|
Romero AH, Aguilera E, Gotopo L, Cabrera G, Dávila B, Cerecetto H. Optimization of the 2-arylquinazoline-4(3 H)one scaffold for a selective and potent antitrypanosomal agent: modulation of the mechanism of action through chemical functionalization. RSC Med Chem 2023; 14:1992-2006. [PMID: 37859724 PMCID: PMC10583831 DOI: 10.1039/d3md00243h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/07/2023] [Indexed: 10/21/2023] Open
Abstract
We sought to identify a potent and selective antitrypanosomal agent through modulation of the mechanism of action of a 2-arylquinazoline scaffold as an antitrypanosomal agent via chemical functionalization at the 4-position. We wished to use the: (i) susceptibility of trypanosomatids towards nitric oxide (NO) and reactive oxygen species (ROS); (ii) capacity of the 4-substituted quinazoline system to act as an antifolate agent. Three quinazolin-based moieties that differed from each other by having at the 4-position key pharmacophores targeting the induction of NO and ROS production were evaluated in vitro against Leishmania infantum and Trypanosoma cruzi parasites and their modes of action were explored. Replacement of an oxygen moiety at the 4-position of the antifolate 2-arylquinazolin-4(3H)one by hydrazinyl and 5-nitrofuryl-hydrazinyl pharmacophores enhanced antitrypanosomatid activity significantly due to promotion of an additional mechanism beyond the antifolate response such as NO or ROS production, respectively. Among the three types of chemical functionalization, the 5-nitrofuryl-hydrazinyl moiety generated the most potent compounds. Compound 3b was a potential candidate thanks to its sub-micromolar response against the promastigotes/amastigotes of L. infantum and epimastigote of T. cruzi, moderate toxicity on macrophages (J774.1), good selectivity index (∼15.1-17.6) and, importantly, non-mutagenic effects. 2-Arylquinazoline could be an attractive platform to design new anti-trypanosomatid agents with the use of key pharmacophores.
Collapse
Affiliation(s)
- Angel H Romero
- Grupo de Química Orgánica Medicinal, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la Republica Igual 4225 11400 Montevideo Uruguay
| | - Elena Aguilera
- Grupo de Química Orgánica Medicinal, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la Republica Igual 4225 11400 Montevideo Uruguay
| | - Lourdes Gotopo
- Laboratorio de Síntesis de Orgánica, Facultad de Ciencias, Universidad Central de Venezuela Los Chaguaramos Caracas 1041-A Venezuela
| | - Gustavo Cabrera
- Laboratorio de Síntesis de Orgánica, Facultad de Ciencias, Universidad Central de Venezuela Los Chaguaramos Caracas 1041-A Venezuela
| | - Belén Dávila
- Grupo de Química Orgánica Medicinal, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la Republica Igual 4225 11400 Montevideo Uruguay
| | - Hugo Cerecetto
- Grupo de Química Orgánica Medicinal, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la Republica Igual 4225 11400 Montevideo Uruguay
- Área de Radiofarmacia, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la Republica Mataojo 42055 11400 Montevideo Uruguay
| |
Collapse
|
29
|
Lourenço EMG, da Silva F, das Neves AR, Bonfá IS, Ferreira AMT, Menezes ACG, da Silva MEC, Dos Santos JT, Martines MAU, Perdomo RT, Toffoli-Kadri MC, G Barbosa E, Saba S, Beatriz A, Rafique J, de Arruda CCP, de Lima DP. Investigation of the Potential Targets behind the Promising and Highly Selective Antileishmanial Action of Synthetic Flavonoid Derivatives. ACS Infect Dis 2023; 9:2048-2061. [PMID: 37772925 DOI: 10.1021/acsinfecdis.3c00336] [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: 09/30/2023]
Abstract
Leishmaniases are among the neglected tropical diseases that still cause devastating health, social, and economic consequences to more than 350 million people worldwide. Despite efforts to combat these vector-borne diseases, their incidence does not decrease. Meanwhile, current antileishmanial drugs are old and highly toxic, and safer presentations are unaffordable to the most severely affected human populations. In a previous study by our research group, we synthesized 17 flavonoid derivatives that demonstrated impressive inhibition capacity against rCPB2.8, rCPB3, and rH84Y. These cysteine proteases are highly expressed in the amastigote stage, the target form of the parasite. However, although these compounds have been already described in the literature, until now, the amastigote effect of any of these molecules has not been proven. In this work, we aimed to deeply analyze the antileishmanial action of this set of synthetic flavonoid derivatives by correlating their ability to inhibit cysteine proteases with the action against the parasite. Among all the synthesized flavonoid derivatives, 11 of them showed high activity against amastigotes of Leishmania amazonensis, also providing safety to mammalian host cells. Furthermore, the high production of nitric oxide by infected cells treated with the most active cysteine protease B (CPB) inhibitors confirms a potential immunomodulatory response of macrophages. Besides, considering flavonoids as multitarget drugs, we also investigated other potential antileishmanial mechanisms. The most active compounds were selected to investigate another potential biological pathway behind their antileishmanial action using flow cytometry analysis. The results confirmed an oxidative stress after 48 h of treatment. These data represent an important step toward the validation of CPB as an antileishmanial target, as well as aiding in new drug discovery studies based on this protease.
Collapse
Affiliation(s)
- Estela M G Lourenço
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| | - Fernanda da Silva
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900, MS, Brazil
| | - Amarith R das Neves
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900, MS, Brazil
| | - Iluska S Bonfá
- Laboratório de Farmacologia e Inflamação, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79074-460 MS, Brazil
| | - Alda Maria T Ferreira
- Laboratório de Imunologia, Biologia Molecular e Bioensaios Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900 MS, Brazil
| | - Adriana C G Menezes
- Biotério Central, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900 MS, Brazil
| | - Maria E C da Silva
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| | - Jéssica T Dos Santos
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| | - Marco A U Martines
- Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| | - Renata T Perdomo
- Laboratório de Biologia Molecular e Cultura de Células, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900 MS, Brazil
| | - Mônica C Toffoli-Kadri
- Laboratório de Farmacologia e Inflamação, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79074-460 MS, Brazil
| | - Euzébio G Barbosa
- Laboratório de Química Farmacêutica Computacional, Departamento de Farmácia, Universidade Federal do Rio Grande do Norte, Natal, 59012-570, RN, Brazil
| | - Sumbal Saba
- Laboratório de Síntese Sustentável e Organocalcogênio - LabSO, Instituto de Química, Universidade Federal de Goiás-UFG, Goiânia, 74690-900 GO, Brazil
| | - Adilson Beatriz
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| | - Jamal Rafique
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
- Laboratório de Síntese Sustentável e Organocalcogênio - LabSO, Instituto de Química, Universidade Federal de Goiás-UFG, Goiânia, 74690-900 GO, Brazil
| | - Carla C P de Arruda
- Laboratório de Parasitologia Humana, Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Campo Grande, 79070-900, MS, Brazil
| | - Dênis P de Lima
- Laboratório de Síntese e Transformação de Moléculas Orgânicas -SINTMOL, Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller, Campo Grande, 79074-460 MS, Brazil
| |
Collapse
|
30
|
Zheng Y, van den Kerkhof M, van der Meer T, Gul S, Kuzikov M, Ellinger B, de Esch IJP, Siderius M, Matheeussen A, Maes L, Sterk GJ, Caljon G, Leurs R. Discovery of 5-Phenylpyrazolopyrimidinone Analogs as Potent Antitrypanosomal Agents with In Vivo Efficacy. J Med Chem 2023; 66:10252-10264. [PMID: 37471520 PMCID: PMC10424178 DOI: 10.1021/acs.jmedchem.3c00161] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Indexed: 07/22/2023]
Abstract
Human African Trypanosomiasis (HAT), caused by Trypanosoma brucei, is one of the neglected tropical diseases with a continuing need for new medication. We here describe the discovery of 5-phenylpyrazolopyrimidinone analogs as a novel series of phenotypic antitrypanosomal agents. The most potent compound, 30 (NPD-2975), has an in vitro IC50 of 70 nM against T. b. brucei with no apparent toxicity against human MRC-5 lung fibroblasts. Showing good physicochemical properties, low toxicity potential, acceptable metabolic stability, and other pharmacokinetic features, 30 was further evaluated in an acute mouse model of T. b. brucei infection. After oral dosing at 50 mg/kg twice per day for five consecutive days, all infected mice were cured. Given its good drug-like properties and high in vivo antitrypanosomal potential, the 5-phenylpyrazolopyrimidinone analog 30 represents a promising lead for future drug development to treat HAT.
Collapse
Affiliation(s)
- Yang Zheng
- Amsterdam
Institute for Molecules, Medicines and Systems, Division of Medicinal
Chemistry, Faculty of Science, Vrije Universiteit
Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Magali van den Kerkhof
- Laboratory
of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Tiffany van der Meer
- Amsterdam
Institute for Molecules, Medicines and Systems, Division of Medicinal
Chemistry, Faculty of Science, Vrije Universiteit
Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Sheraz Gul
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
- Fraunhofer
Cluster of Excellence for Immune-Mediated Diseases CIMD, 22525 Hamburg, Germany
| | - Maria Kuzikov
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
- Fraunhofer
Cluster of Excellence for Immune-Mediated Diseases CIMD, 22525 Hamburg, Germany
| | - Bernhard Ellinger
- Fraunhofer
Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
- Fraunhofer
Cluster of Excellence for Immune-Mediated Diseases CIMD, 22525 Hamburg, Germany
| | - Iwan J. P. de Esch
- Amsterdam
Institute for Molecules, Medicines and Systems, Division of Medicinal
Chemistry, Faculty of Science, Vrije Universiteit
Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Marco Siderius
- Amsterdam
Institute for Molecules, Medicines and Systems, Division of Medicinal
Chemistry, Faculty of Science, Vrije Universiteit
Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - An Matheeussen
- Laboratory
of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Louis Maes
- Laboratory
of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Geert Jan Sterk
- Amsterdam
Institute for Molecules, Medicines and Systems, Division of Medicinal
Chemistry, Faculty of Science, Vrije Universiteit
Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Guy Caljon
- Laboratory
of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Rob Leurs
- Amsterdam
Institute for Molecules, Medicines and Systems, Division of Medicinal
Chemistry, Faculty of Science, Vrije Universiteit
Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
31
|
Pal R, Teli G, Akhtar MJ, Matada GSP. The role of natural anti-parasitic guided development of synthetic drugs for leishmaniasis. Eur J Med Chem 2023; 258:115609. [PMID: 37421889 DOI: 10.1016/j.ejmech.2023.115609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/10/2023]
Abstract
Leishmaniasis is a parasitic disease and categorised as a neglected tropical disease (NTD). Each year, between 70,0000 and 1 million new cases are believed to occur. There are approximately 90 sandfly species which can spread the Leishmania parasites (over 20 species) causing 20,000 to 30,000 death per year. Currently, leishmaniasis has no specific therapeutic treatment available. The prescribed drugs with several drawbacks including high cost, challenging administration, toxicity, and drug resistance led to search for the alternative treatment with less toxicity and selectivity. Introducing the molecular features like that of phytoconstituents for the search of compounds with less toxicity is another promising approach. The current review classifies the synthetic compounds according to the core rings present in the natural phytochemicals for the development of antileishmanial agents (2020-2022). Considering the toxicity and limitations of synthetic analogues, natural compounds are at the higher notch in terms of effectiveness and safety. Synthesized compounds of chalcones (Compound 8; IC50: 0.03 μM, 4.7 folds more potent than Amphotericin B; IC50: 0.14 μM), pyrimidine (compound 56; against L. tropica; 0.04 μM and L. infantum; 0.042 μM as compared to glucantime: L. tropica; 8.17 μM and L. infantum; 8.42 μM), quinazoline and (compound 72; 0.021 μM, 150 times more potent than miltefosine). The targeted delivery against DHFR have been demonstrated by one of the pyrimidine compounds 62 with an IC50 value of 0.10 μM against L. major as compared to the standard trimethoprim (IC50: 20 μM). The review covers the medicinal importance of antileishmanial agents from synthetic and natural sources such as chalcone, pyrazole, coumarins, steroids, and alkaloidal-containing drugs (indole, quinolines, pyridine, pyrimidine, carbolines, pyrrole, aurones, and quinazolines). The efforts of introducing the core rings present in the natural phytoconstituents as antileishmanial in the synthetic compounds are discussed with their structural activity relationship. The perspective will support the medicinal chemists in refining and directing the development of novel molecules phytochemicals-based antileishmanial agents.
Collapse
Affiliation(s)
- Rohit Pal
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| | - Ghanshyam Teli
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| | - Md Jawaid Akhtar
- Department of Pharmaceutical Chemistry, National University of Science and Technology, PO 620, PC 130, Azaiba Bousher, Muscat, Sultanate of Oman
| | - Gurubasavaraja Swamy Purawarga Matada
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| |
Collapse
|
32
|
Kim HS, Ortiz D, Kadayat TM, Fargo CM, Hammill JT, Chen Y, Rice AL, Begley KL, Shoeran G, Pistel W, Yates PA, Sanchez MA, Landfear SM, Guy RK. Optimization of Orally Bioavailable Antileishmanial 2,4,5-Trisubstituted Benzamides. J Med Chem 2023; 66:7374-7386. [PMID: 37216489 PMCID: PMC10259451 DOI: 10.1021/acs.jmedchem.3c00056] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Indexed: 05/24/2023]
Abstract
Leishmaniasis, a neglected tropical disease caused by Leishmania species parasites, annually affects over 1 million individuals worldwide. Treatment options for leishmaniasis are limited due to high cost, severe adverse effects, poor efficacy, difficulty of use, and emerging drug resistance to all approved therapies. We discovered 2,4,5-trisubstituted benzamides (4) that possess potent antileishmanial activity but poor aqueous solubility. Herein, we disclose our optimization of the physicochemical and metabolic properties of 2,4,5-trisubstituted benzamide that retains potency. Extensive structure-activity and structure-property relationship studies allowed selection of early leads with suitable potency, microsomal stability, and improved solubility for progression. Early lead 79 exhibited an 80% oral bioavailability and potently blocked proliferation of Leishmania in murine models. These benzamide early leads are suitable for development as orally available antileishmanial drugs.
Collapse
Affiliation(s)
- Ho Shin Kim
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509 United States
| | - Diana Ortiz
- Department
of Molecular Microbiology & Immunology, Oregon Health and Science University, Portland, Oregon 97239 United States
| | - Tara Man Kadayat
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509 United States
| | - Corinne M. Fargo
- Department
of Molecular Microbiology & Immunology, Oregon Health and Science University, Portland, Oregon 97239 United States
- Department
of Chemical Physiology & Biochemistry, Oregon Health and Science University, Portland, Oregon 97239 United States
| | - Jared T. Hammill
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509 United States
| | - Yizhe Chen
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509 United States
| | - Amy L. Rice
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509 United States
| | - Kristin L. Begley
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509 United States
| | - Gaurav Shoeran
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509 United States
| | - William Pistel
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509 United States
| | - Phillip A. Yates
- Department
of Chemical Physiology & Biochemistry, Oregon Health and Science University, Portland, Oregon 97239 United States
| | - Marco A. Sanchez
- Department
of Molecular Microbiology & Immunology, Oregon Health and Science University, Portland, Oregon 97239 United States
| | - Scott M. Landfear
- Department
of Molecular Microbiology & Immunology, Oregon Health and Science University, Portland, Oregon 97239 United States
- Department
of Chemical Physiology & Biochemistry, Oregon Health and Science University, Portland, Oregon 97239 United States
| | - R. Kiplin Guy
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509 United States
| |
Collapse
|
33
|
Vásquez-Ocmín PG, Cojean S, Roumy V, Marti G, Pomel S, Gadea A, Leblanc K, Dennemont I, Ruiz-Vásquez L, Ricopa Cotrina H, Ruiz Mesia W, Bertani S, Ruiz Mesia L, Maciuk A. Deciphering anti-infectious compounds from Peruvian medicinal Cordoncillos extract library through multiplexed assays and chemical profiling. Front Pharmacol 2023; 14:1100542. [PMID: 37342590 PMCID: PMC10278888 DOI: 10.3389/fphar.2023.1100542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/04/2023] [Indexed: 06/23/2023] Open
Abstract
High prevalence of parasitic or bacterial infectious diseases in some world areas is due to multiple reasons, including a lack of an appropriate health policy, challenging logistics and poverty. The support to research and development of new medicines to fight infectious diseases is one of the sustainable development goals promoted by World Health Organization (WHO). In this sense, the traditional medicinal knowledge substantiated by ethnopharmacology is a valuable starting point for drug discovery. This work aims at the scientific validation of the traditional use of Piper species ("Cordoncillos") as firsthand anti-infectious medicines. For this purpose, we adapted a computational statistical model to correlate the LCMS chemical profiles of 54 extracts from 19 Piper species to their corresponding anti-infectious assay results based on 37 microbial or parasites strains. We mainly identified two groups of bioactive compounds (called features as they are considered at the analytical level and are not formally isolated). Group 1 is composed of 11 features being highly correlated to an inhibiting activity on 21 bacteria (principally Gram-positive strains), one fungus (C. albicans), and one parasite (Trypanosoma brucei gambiense). The group 2 is composed of 9 features having a clear selectivity on Leishmania (all strains, both axenic and intramacrophagic). Bioactive features in group 1 were identified principally in the extracts of Piper strigosum and P. xanthostachyum. In group 2, bioactive features were distributed in the extracts of 14 Piper species. This multiplexed approach provided a broad picture of the metabolome as well as a map of compounds putatively associated to bioactivity. To our knowledge, the implementation of this type of metabolomics tools aimed at identifying bioactive compounds has not been used so far.
Collapse
Affiliation(s)
| | - Sandrine Cojean
- Université Paris-Saclay, CNRS, BioCIS, Orsay, France
- CNR Du Paludisme, AP-HP, Hôpital Bichat–Claude Bernard, Paris, France
| | - Vincent Roumy
- Joint Research Unit 1158 BioEcoAgro, University Lille, JUNIA, INRAE, University Liège, UPJV, University Artois, ULCO, VilleneuveD’Ascq, France
| | - Guillaume Marti
- Laboratoire de Recherche en Sciences Végétales (UMR 5546), CNRS, Université de Toulouse, Toulouse, France
- MetaboHUB, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | | | - Alice Gadea
- UMR152 PHARMADEV, IRD, UPS, Université de Toulouse, Toulouse, France
| | | | | | - Liliana Ruiz-Vásquez
- Facultad de Farmacia y Bioquímica, Universidad Nacional de la Amazonía Peruana (UNAP), Iquitos, Peru
- Centro de Investigación de Recursos Naturales, Universidad Nacional de la Amazonía Peruana (UNAP), Iquitos, Peru
| | - Hivelli Ricopa Cotrina
- Centro de Investigación de Recursos Naturales, Universidad Nacional de la Amazonía Peruana (UNAP), Iquitos, Peru
| | - Wilfredo Ruiz Mesia
- Facultad de Ingeniería Química, Universidad Nacional de la Amazonía Peruana (UNAP), Iquitos, Peru
| | - Stéphane Bertani
- UMR152 PHARMADEV, IRD, UPS, Université de Toulouse, Toulouse, France
- International Joint Laboratory of Molecular Anthropological Oncology (LOAM), National Cancer Institute, Lima, Perú
| | - Lastenia Ruiz Mesia
- Centro de Investigación de Recursos Naturales, Universidad Nacional de la Amazonía Peruana (UNAP), Iquitos, Peru
- Facultad de Ingeniería Química, Universidad Nacional de la Amazonía Peruana (UNAP), Iquitos, Peru
| | | |
Collapse
|
34
|
Afonso RC, Yien RMK, de Siqueira LBDO, Simas NK, Dos Santos Matos AP, Ricci-Júnior E. Promising natural products for the treatment of cutaneous leishmaniasis: A review of in vitro and in vivo studies. Exp Parasitol 2023; 251:108554. [PMID: 37268108 DOI: 10.1016/j.exppara.2023.108554] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/04/2023] [Accepted: 05/24/2023] [Indexed: 06/04/2023]
Abstract
Although there are available treatments for cutaneous leishmaniasis (CL), the drugs used are far from ideal, toxic, and costly, in addition to the challenge faced by the development of resistance. Plants have been used as a source of natural compounds with antileishmanial action. However, few have reached the market and become phytomedicines with registration in regulatory agencies. Difficulties related to the extraction, purification, chemical identification, efficacy, safety, and production in sufficient quantity for clinical studies, hinder the emergence of new effective phytomedicines against leishmaniasis. Despite the difficulties reported, the major research centers in the world see that natural products are a trend concerning the treatment of leishmaniasis. The present work consists of a literature review of articles with in vivo studies, covering the period from January 2011 to December 2022, providing an overview of promising natural products for CL treatment. The papers show encouraging antileishmanial action of natural compounds with reduced parasite load and lesion size in animal models, suggesting new strategies for the treatment of the disease. The results reported in this review show advances in using natural products as safe and effective formulations, which can stimulate clinical studies to establish clinical therapy. In conclusion, the information in this review article serves as a preliminary basis for establishing a therapeutic protocol for future clinical trials that can validate the safety and efficacy of natural compounds, providing the development of affordable and safe phytomedicines for the treatment of CL.
Collapse
Affiliation(s)
- Rhuane Coutinho Afonso
- Galenic Development Laboratory (LADEG), Department of Drugs and Medicines, Faculty of Pharmacy, Federal University of Rio de Janeiro, RJ, Brazil
| | - Raíssa Mara Kao Yien
- Galenic Development Laboratory (LADEG), Department of Drugs and Medicines, Faculty of Pharmacy, Federal University of Rio de Janeiro, RJ, Brazil; Laboratory of Natural Products and Biological Assays, Department of Natural Products and Food, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Naomi Kato Simas
- Laboratory of Natural Products and Biological Assays, Department of Natural Products and Food, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ana Paula Dos Santos Matos
- Galenic Development Laboratory (LADEG), Department of Drugs and Medicines, Faculty of Pharmacy, Federal University of Rio de Janeiro, RJ, Brazil
| | - Eduardo Ricci-Júnior
- Galenic Development Laboratory (LADEG), Department of Drugs and Medicines, Faculty of Pharmacy, Federal University of Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
35
|
Jacques Dit Lapierre TJW, Cruz MGFDML, Brito NPF, Resende DDM, Souza FDO, Pilau EJ, da Silva MFB, Neves BJ, Murta SMF, Rezende Júnior CDO. Hit-to-lead optimization of a pyrazinylpiperazine series against Leishmania infantum and Leishmania braziliensis. Eur J Med Chem 2023; 256:115445. [PMID: 37156183 DOI: 10.1016/j.ejmech.2023.115445] [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: 01/26/2023] [Revised: 04/17/2023] [Accepted: 05/01/2023] [Indexed: 05/10/2023]
Abstract
An early hit-to-lead optimization of a novel pyrazinylpiperazine series against L. infantum and L. braziliensis has been performed after an extensive SAR focusing on the benzoyl fragment of hit (4). Deletion of the meta-Cl of (4) led to the obtention of the para-hydroxyl derivative (12), on which the design of most monosubstituted derivatives of the SAR was based. Further optimization of the series, involving disubstituted benzoyl fragments and the hydroxyl substituent of (12), allowed the obtention of a total of 15 compounds with increased antileishmanial potency (IC50 < 10 μM), nine of which displayed activity in the low micromolar range (IC50 < 5 μM). This optimization ultimately identified the ortho, meta-dihydroxyl derivative (46) as an early lead for this series (IC50 (L. infantum) = 2.8 μM, IC50 (L. braziliensis) = 0.2 μM). Additional assessment of some selected compounds against other trypanosomatid parasites revealed that this series is selective towards Leishmania parasites, and in silico ADMET predictions revealed satisfactory profiles for these compounds, allowing further lead optimization of the pyrazinylpiperazine class against Leishmania.
Collapse
Affiliation(s)
| | | | - Nícolas Peterson Ferreira Brito
- Laboratório de Síntese de Candidatos a Fármacos, Instituto de Química, Universidade Federal de Uberlândia (UFU), Uberlândia, MG, 38400-902, Brazil
| | - Daniela de Melo Resende
- Grupo de Genômica Funcional de Parasitos, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ Minas), Belo Horizonte, MG, 30190-002, Brazil
| | - Felipe de Oliveira Souza
- Laboratório de Biomoléculas e Espectrometria de Massas (LaBioMass), Universidade Estadual de Maringá (UEM), Maringá, PR, 807020-900, Brazil
| | - Eduardo Jorge Pilau
- Laboratório de Biomoléculas e Espectrometria de Massas (LaBioMass), Universidade Estadual de Maringá (UEM), Maringá, PR, 807020-900, Brazil
| | - Meryck Felipe Brito da Silva
- Laboratory of Cheminformatics (LabChem), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, 74605-170, Brazil
| | - Bruno Junior Neves
- Laboratory of Cheminformatics (LabChem), Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, 74605-170, Brazil
| | - Silvane Maria Fonseca Murta
- Grupo de Genômica Funcional de Parasitos, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ Minas), Belo Horizonte, MG, 30190-002, Brazil
| | - Celso de Oliveira Rezende Júnior
- Laboratório de Síntese de Candidatos a Fármacos, Instituto de Química, Universidade Federal de Uberlândia (UFU), Uberlândia, MG, 38400-902, Brazil.
| |
Collapse
|
36
|
Bhakta S, Bhattacharya A. In silico evolutionary and structural analysis of cAMP response proteins (CARPs) from Leishmania major. Arch Microbiol 2023; 205:125. [PMID: 36941487 DOI: 10.1007/s00203-023-03463-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/23/2023]
Abstract
With unidentified chemical triggers and novel-effectors, cAMP signaling is broadly noncanonical in kinetoplastida parasites. Though novel protein kinase A regulatory subunits (PKAR) have been identified earlier, cAMP Response Proteins (CARPs) have been identified as a unique and definite cAMP effector of trypanosomatids. CARP1-CARP4 emerged as critical regulatory components of cAMP signaling pathway in Trypanosoma with evidences that CARP3 can directly interact with a flagellar adenylate cyclase (AC). CARP-mediated regulations, identified so far, reflects the mechanistic diversity of cAMP signaling. Albeit the function of the orthologous is not yet delineated, in kinetoplastids like Leishmania, presence of CARP1, 2 and 4 orthologues suggests existence of conserved effector mechanisms. Targeting CARP orthologues in Leishmania, a comprehensive evolutionary analysis of CARPs have been aimed in this study which revealed phylogenetic relationship, codon adaptation and structural heterogeneity among the orthologues, warranting functional analysis in future to explore their involvement in infectivity.
Collapse
Affiliation(s)
- Swarnav Bhakta
- Department of Biotechnology, Adamas University, Barasat-Barrackpore Rd., Kolkata, 700126, India
| | - Arijit Bhattacharya
- Department of Microbiology, Adamas University, Barasat-Barrackpore Rd., Kolkata, 700126, India.
| |
Collapse
|
37
|
Ketema T, Tadele M, Gebrie Z, Makonnen E, Hailu A, Abay SM. In vitro Anti-Leishmanial Activities of Methanol Extract of Brucea antidysenterica J.F. Mill Seeds and Its Solvent Fractions. J Exp Pharmacol 2023; 15:123-135. [PMID: 36937078 PMCID: PMC10022440 DOI: 10.2147/jep.s397352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/02/2023] [Indexed: 03/15/2023] Open
Abstract
Introduction Leishmaniasis is one of the neglected tropical diseases, threatening lives of about 350 million people globally. Brucea antidysenterica seeds are used for the treatment of cutaneous leishmaniasis in the traditional medicine in Ethiopia. Objective This study aimed to evaluate Brucea antidysenterica seeds' anti-leishmanial activity in vitro. Methods The crude (80% methanol) extract of Brucea antidysenterica seeds and its fractions were evaluated for their anti-leishmanial activities against promastigotes and intracellular amastigotes of Leishmania donovani and Leishmania aethiopica, and for their cytotoxic effects against mammalian cells. The quantitative estimations of total phenolic compounds (TPCs), flavonoids (TFCs) and alkaloids (TACs) were determined, spectrophotometrically. Median inhibitory concentration (IC50) and median cytotoxic concentration (CC50) of the extract and its solvent fractions were calculated using GraphPad Prism 9.1.0 computer software. Data was presented as mean ± standard error of the mean (SEM). Results The crude extract and its hexane, ethyl acetate and butanol fractions showed anti-leishmanial activities, with IC50 values of 4.14-60.12 µg/mL against promastigotes, and 6.16-40.12 µg/mL against amastigotes of both Leishmania species. They showed moderate cytotoxicity against Vero cell lines and peritoneal mice macrophages, with CC50 values of 100-500 µg/mL, but >1600 µg/mL against red blood cells. Selectivity indices ranged from 7.97 to 30.97. The crude extract, and its ethyl acetate and hexane fractions possessed 54.78-127.72 mg of gallic acid equivalent TPC, 18.30-79.21 mg of quercetin equivalent TFC, and 27.62-97.22 mg of atropine equivalent TAC per gram of extracts. Conclusion The seeds of the plant possessed anti-leishmanial activities against L. aethiopica and L. donovani that might provide a scientific justification for its use in the treatment of leishmaniasis by traditional healers. Future works are recommended to isolate, purify and identify the possible secondary metabolites attributed to the anti-leishmanial activity.
Collapse
Affiliation(s)
- Tasisa Ketema
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia
| | - Markos Tadele
- Animal Health Research Program, Ethiopian Institute of Agricultural Research, Holetta, Ethiopia
| | - Zewdie Gebrie
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia
| | - Eyasu Makonnen
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Asrat Hailu
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT Africa), College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
- Department of Microbiology, Immunology and Parasitology, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Solomon M Abay
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
38
|
Ndlovu K, Kannigadu C, Aucamp J, van Rensburg HDJ, N'Da DD. Exploration of ethylene glycol linked nitrofurantoin derivatives against Leishmania: Synthesis and in vitro activity. Arch Pharm (Weinheim) 2023; 356:e2200529. [PMID: 36759973 DOI: 10.1002/ardp.202200529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/27/2022] [Accepted: 12/30/2022] [Indexed: 02/11/2023]
Abstract
Leishmaniasis is a neglected tropical disease that is caused by the Leishmania parasite. It is estimated that there are more than 350 million people at risk of infection annually. Current treatments that are in clinical use are expensive, have toxic side effects, and are facing parasitic resistance. Therefore, new drugs are urgently required. In the quest for new, safe, and cost-effective drugs, a series of novel ethylene glycol derivatives of nitrofurantoin was synthesised and the in vitro antileishmanial efficacy of the compounds tested against Leishmania donovani and Leishmania major strains. Arylated ethylene glycol derivatives were found to be the most potent, with submicromolar activity up to 294-fold greater than the parent compound nitrofurantoin. Analogues 2j and 2k had the best antipromastigote activities with submicromolar IC50 values against L. major IR-173 and antimonial-resistant L. donovani 9515 strains.
Collapse
Affiliation(s)
- Keitumetsi Ndlovu
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Christina Kannigadu
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | - Janine Aucamp
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | | | - David D N'Da
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
39
|
Gupta D, Singh PK, Yadav PK, Narender T, Patil UK, Jain SK, Chourasia MK. Emerging strategies and challenges of molecular therapeutics in antileishmanial drug development. Int Immunopharmacol 2023; 115:109649. [PMID: 36603357 DOI: 10.1016/j.intimp.2022.109649] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/16/2022] [Accepted: 12/24/2022] [Indexed: 01/05/2023]
Abstract
Molecular therapy refers to targeted therapies based on molecules which have been intelligently directed towards specific biomolecular structures and include small molecule drugs, monoclonal antibodies, proteins and peptides, DNA or RNA-based strategies, targeted chemotherapy and nanomedicines. Molecular therapy is emerging as the most effective strategy to combat the present challenges of life-threatening visceral leishmaniasis, where the successful human vaccine is currently unavailable. Moreover, current chemotherapy-based strategies are associated with the issues of ineffective targeting, unavoidable toxicities, invasive therapies, prolonged treatment, high treatment costs and the development of drug-resistant strains. Thus, the rational approach to antileishmanial drug development primarily demands critical exploration and exploitation of biochemical differences between host and parasite biology, immunocharacteristics of parasite homing, and host-parasite interactions at the molecular/cellular level. Following this, the novel technology-based designing and development of host and/or parasite-targeted therapeutics having leishmanicidal and immunomodulatory activity is utmost essential to improve treatment efficacy. Thus, the present review is focused on immunological and molecular checkpoint targets in host-pathogen interaction, and molecular therapeutic prospects for Leishmania intervention, and the challenges ahead.
Collapse
Affiliation(s)
- Deepak Gupta
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India; Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Pankaj K Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India
| | - Pavan K Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Tadigoppula Narender
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Umesh K Patil
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India
| | - Sanjay K Jain
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India.
| |
Collapse
|
40
|
Tawaraishi T, Ochida A, Akao Y, Itono S, Kamaura M, Akther T, Shimada M, Canan S, Chowdhury S, Cao Y, Condroski K, Engkvist O, Francisco A, Ghosh S, Kaki R, Kelly JM, Kimura C, Kogej T, Nagaoka K, Naito A, Pairaudeau G, Radu C, Roberts I, Shum D, Watanabe NA, Xie H, Yonezawa S, Yoshida O, Yoshida R, Mowbray C, Perry B. Collaborative Virtual Screening Identifies a 2-Aryl-4-aminoquinazoline Series with Efficacy in an In Vivo Model of Trypanosoma cruzi Infection. J Med Chem 2023; 66:1221-1238. [PMID: 36607408 PMCID: PMC9884087 DOI: 10.1021/acs.jmedchem.2c00775] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Probing multiple proprietary pharmaceutical libraries in parallel via virtual screening allowed rapid expansion of the structure-activity relationship (SAR) around hit compounds with moderate efficacy against Trypanosoma cruzi, the causative agent of Chagas Disease. A potency-improving scaffold hop, followed by elaboration of the SAR via design guided by the output of the phenotypic virtual screening efforts, identified two promising hit compounds 54 and 85, which were profiled further in pharmacokinetic studies and in an in vivo model of T. cruzi infection. Compound 85 demonstrated clear reduction of parasitemia in the in vivo setting, confirming the interest in this series of 2-(pyridin-2-yl)quinazolines as potential anti-trypanosome treatments.
Collapse
Affiliation(s)
- Taisuke Tawaraishi
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Atsuko Ochida
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yuichiro Akao
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Sachiko Itono
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masahiro Kamaura
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Thamina Akther
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Mitsuyuki Shimada
- Takeda
Pharmaceutical Company Limited, 26-1 Muraoka-Higashi 2-chrome, Fujisawa, Kanagawa 251-8555, Japan
| | - Stacie Canan
- Celgene
Corporation, Celgene Global Health, 10300 Campus Point Drive, San Diego, California 92121, United States
| | - Sanjoy Chowdhury
- TCG
Lifesciences, Plot No-7,
Salt Lake Electronics Complex, BN Block, Sector V, Kolkata 700091, India
| | - Yafeng Cao
- WuXi
AppTec Company Ltd., 666 Gaoxin Road, East Lake High-Tech Development Zone, Wuhan 430075, People’s Republic of China
| | - Kevin Condroski
- Celgene
Corporation, Celgene Global Health, 10300 Campus Point Drive, San Diego, California 92121, United States
| | - Ola Engkvist
- AstraZeneca
Discovery Sciences, R&D, Pepparedsleden 1, 431 50 Mölndal, Sweden
| | - Amanda Francisco
- London School
of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K.
| | - Sunil Ghosh
- TCG
Lifesciences, Plot No-7,
Salt Lake Electronics Complex, BN Block, Sector V, Kolkata 700091, India
| | - Rina Kaki
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - John M. Kelly
- London School
of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, U.K.
| | - Chiaki Kimura
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Thierry Kogej
- AstraZeneca
Discovery Sciences, R&D, Pepparedsleden 1, 431 50 Mölndal, Sweden
| | - Kazuya Nagaoka
- Eisai
Co., Ltd, 1-3, Tokodai
5-chome, Tsukuba, Ibaraki 300-2635, Japan
| | - Akira Naito
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Garry Pairaudeau
- AstraZeneca,
Discovery Sciences, R&D, The Darwin Building, 310 Milton Road, Milton, Cambridge CB4 0WG, U.K.
| | - Constantin Radu
- Institut
Pasteur Korea, 16, Daewangpangyo-ro
712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Ieuan Roberts
- AstraZeneca,
Discovery Sciences, R&D, The Darwin Building, 310 Milton Road, Milton, Cambridge CB4 0WG, U.K.
| | - David Shum
- Institut
Pasteur Korea, 16, Daewangpangyo-ro
712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Nao-aki Watanabe
- Eisai
Co., Ltd, 1-3, Tokodai
5-chome, Tsukuba, Ibaraki 300-2635, Japan
| | - Huanxu Xie
- WuXi
AppTec Company Ltd., 666 Gaoxin Road, East Lake High-Tech Development Zone, Wuhan 430075, People’s Republic of China
| | - Shuji Yonezawa
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Osamu Yoshida
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Ryu Yoshida
- Shionogi
& Co., Ltd, 3-1-1,
Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Charles Mowbray
- Drugs for Neglected
Diseases Initiative, 15 Chemin Camille Vidart, Geneva 1202, Switzerland
| | - Benjamin Perry
- Drugs for Neglected
Diseases Initiative, 15 Chemin Camille Vidart, Geneva 1202, Switzerland,
| |
Collapse
|
41
|
Gopu B, Kour P, Pandian R, Singh K. Insights into the drug screening approaches in leishmaniasis. Int Immunopharmacol 2023; 114:109591. [PMID: 36700771 DOI: 10.1016/j.intimp.2022.109591] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/25/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
Leishmaniasis, a tropically neglected disease, is responsible for the high mortality and morbidity ratio in poverty-stricken areas. Currently, no vaccine is available for the complete cure of the disease. Current chemotherapeutic regimens face the limitations of drug resistance and toxicity concerns indicating a great need to develop better chemotherapeutic leads that are orally administrable, potent, non-toxic, and cost-effective. The anti-leishmanial drug discovery process accelerated the desire for large-scale drug screening assays and high-throughput screening (HTS) technology to identify new chemo-types that can be used as potential drug molecules to control infection. Using the HTS approach, about one million compounds can be screened daily within the shortest possible time for biological activity using automation tools, miniaturized assay formats, and large-scale data analysis. Classical and modern in vitro screening assays have led to the progression of active compounds further to ex vivo and in vivo studies. In the present review, we emphasized on the HTS approaches employed in the leishmanial drug discovery program. Recent in vitro screening assays are widely explored to discover new chemical scaffolds. Developing appropriate experimental animal models and their related techniques is necessary to understand the pathophysiological processes and disease host responses, paving the way for unraveling novel therapies against leishmaniasis.
Collapse
Affiliation(s)
- Boobalan Gopu
- Animal House Facility, Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Parampreet Kour
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Ramajayan Pandian
- Animal House Facility, Pharmacology Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kuljit Singh
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
42
|
Rivas F, Del Mármol C, Scalese G, Pérez-Díaz L, Machado I, Blacque O, Medeiros A, Comini M, Gambino D. New multifunctional Ru(II) organometallic compounds show activity against Trypanosoma brucei and Leishmania infantum. J Inorg Biochem 2022; 237:112016. [PMID: 36244312 DOI: 10.1016/j.jinorgbio.2022.112016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022]
Abstract
Human African trypanosomiasis (sleeping sickness) and leishmaniasis are prevalent zoonotic diseases caused by genomically related trypanosomatid protozoan parasites (Trypanosoma brucei and Leishmania spp). Additionally, both are co-endemic in certain regions of the world. Only a small number of old drugs exist for their treatment, with most of them sharing poor safety, efficacy, and pharmacokinetic profiles. In this work, new multifunctional Ru(II) ferrocenyl compounds were rationally designed as potential agents against these trypanosomatid parasites by including in a single molecule 1,1'-bis(diphenylphosphino)ferrocene (dppf) and two bioactive bidentate ligands: 8-hydroxyquinoline derivatives (8HQs) and polypyridyl ligands (NN). Three [Ru(8HQs)(dppf)(NN)](PF6) compounds were synthesized and fully characterized. They showed in vitro activity on bloodstream Trypanosoma brucei (IC50 140-310 nM) and on Leishmania infantum promastigotes (IC50 3.0-4.8 μM). The compounds showed good selectivity towards T. brucei in respect to J774 murine macrophages as mammalian cell model (SI 15-38). Changing hexafluorophosphate counterion by chloride led to a three-fold increase in activity on both parasites and to a two to three-fold increase in selectivity towards the pathogens. The compounds affect in vitro at least the targets of the individual bioactive moieties included in the new chemical entities: DNA and generation of ROS. The compounds are stable in solution and are more lipophilic than the free bioactive ligands. No clear correlation between lipophilicity, interaction with DNA or generation of ROS and activity was detected, which agrees with their overall similar anti-trypanosoma potency and selectivity. These compounds are promising candidates for further drug development.
Collapse
Affiliation(s)
- Feriannys Rivas
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay; Programa de Posgrado en Química, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Carolina Del Mármol
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Gonzalo Scalese
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Leticia Pérez-Díaz
- Laboratorio de Interacciones Moleculares, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Ignacio Machado
- Área Química Analítica, Universidad de la República, Montevideo, Uruguay
| | - Olivier Blacque
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Andrea Medeiros
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Montevideo, Uruguay; Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Marcelo Comini
- Group Redox Biology of Trypanosomes, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Dinorah Gambino
- Área Química Inorgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
43
|
Feng M, Jin Y, Yang S, Joachim AM, Ning Y, Mori-Quiroz LM, Fromm J, Perera C, Zhang K, Werbovetz KA, Wang MZ. Sterol profiling of Leishmania parasites using a new HPLC-tandem mass spectrometry-based method and antifungal azoles as chemical probes reveals a key intermediate sterol that supports a branched ergosterol biosynthetic pathway. Int J Parasitol Drugs Drug Resist 2022; 20:27-42. [PMID: 35994895 PMCID: PMC9418051 DOI: 10.1016/j.ijpddr.2022.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/27/2022] [Accepted: 07/31/2022] [Indexed: 12/14/2022]
Abstract
Human leishmaniasis is an infectious disease caused by Leishmania protozoan parasites. Current chemotherapeutic options against the deadly disease have significant limitations. The ergosterol biosynthetic pathway has been identified as a drug target in Leishmania. However, remarkable differences in the efficacy of antifungal azoles that inhibit ergosterol biosynthesis have been reported for the treatment of leishmaniasis. To better understand the sterol biosynthetic pathway in Leishmania and elucidate the mechanism underlying the differential efficacy of antifungal azoles, we developed a new LC-MS/MS method to study sterol profiles in promastigotes of three Leishmania species, including two L. donovani, one L. major and one L. tarentolae strains. A combination of distinct precursor ion masses and LC retention times allowed for specific detection of sixteen intermediate sterols between lanosterol and ergosterol using the newly developed LC-MS/MS method. Although both posaconazole and fluconazole are known inhibitors of fungal lanosterol 14α-demethylase (CYP51), only posaconazole led to a substantial accumulation of lanosterol in azole-treated L. donovani promastigotes. Furthermore, a key intermediate sterol accumulated by 40- and 7-fold when these parasites were treated with posaconazole and fluconazole, respectively, which was determined as 4α,14α-dimethylzymosterol by high resolution mass spectrometry and NMR spectroscopy. The identification of 4α,14α-dimethylzymosterol supports a branched ergosterol biosynthetic pathway in Leishmania, where lanosterol C4- and C14-demethylation reactions occur in parallel rather than sequentially. Our results suggest that selective inhibition of leishmanial CYP51 is insufficient to effectively prevent parasite growth and dual inhibitors of both CYP51 and the unknown sterol C4-demethylase may be required for optimal antiparasitic effect.
Collapse
Affiliation(s)
- Mei Feng
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS, USA
| | - Yiru Jin
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS, USA
| | - Sihyung Yang
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS, USA
| | - Arline M Joachim
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Yu Ning
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, 79409, USA
| | - Luis M Mori-Quiroz
- Synthetic Chemical Biology Core Laboratory, The University of Kansas, Lawrence, KS, USA
| | - Jacob Fromm
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS, USA
| | - Chamani Perera
- Synthetic Chemical Biology Core Laboratory, The University of Kansas, Lawrence, KS, USA
| | - Kai Zhang
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, 79409, USA
| | - Karl A Werbovetz
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Michael Zhuo Wang
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
44
|
Majhi B, Parwez A, Palit S, Dutta S. One-Pot Cascade Annulation-Triggered Synthesis of N-6-Substituted Norcryptotackieine Alkaloids and Evaluation of Their Antileishmanial Activities. J Org Chem 2022; 87:14695-14705. [PMID: 36223596 DOI: 10.1021/acs.joc.2c02007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Norcryptotackieine or 6H-indolo[2,3-b]quinoline is an indoloquinoline class of alkaloid isolated from Cryptolepis sanguinolenta that is traditionally used for antimalarial therapy. Additional structural tuning can extend the therapeutic potency of these indoloquinolines as antileishmanial drug leads. Synthesis of N-6-functionalized norcryptotackieines suffers from the necessity of complex pre-synthesized starting materials, restricted scope of functionalization, or tedious processes. Consequently, a straightforward synthetic procedure for accessing non-natural N-6-functionalized 6H-indolo[2,3-b]quinolines with potent antileishmanial activities is highly sought-after. Herein, we report a two-step one-pot synthesis of N-6-functionalized norcryptotackieine through a Pd-catalyzed double annulation reaction of commercially available amphipathic amines, 2-iodobenzyl cyanide, and differently functionalized 2-bromobenzaldehydes. The reported procedure allows a broad flexibility of substitution at the N-6 position and access to diversified scaffolds, including two natural products norcryptotackieine and neocryptolepine. Interestingly, 6d showed potent antileishmanial activities by causing disruption in the cytoskeletal structure and apoptotic-mediated death of parasites. Together, our work manifests the shortest route to N-6-substituted norcryptotackieine-derived antileishmanial agents.
Collapse
Affiliation(s)
- Bhim Majhi
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata 700032, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Aymen Parwez
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata 700032, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Subhadeep Palit
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata 700032, India
| | - Sanjay Dutta
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata 700032, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
45
|
Mert U, Müftüoğlu C, Erdem S, Sadıqova A, Toz S, Ozbel Y, Caner A. The Effect of BTK Inhibitor Ibrutinib on Leishmania infantum Infection In Vitro. Acta Parasitol 2022; 67:1732-1739. [PMID: 36260194 DOI: 10.1007/s11686-022-00630-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 10/10/2022] [Indexed: 11/01/2022]
Abstract
PURPOSE Leishmaniasis is a neglected infectious disease affecting millions of people worldwide. Visceral leishmaniasis (VL), caused by Leishmania infantum and Leishmania donovani, is one of the main clinical forms of the disease and fatal if not treated promptly and properly. Despite being available for the last 70 years, current drugs used in the treatment of leishmaniasis have serious problems as they have high toxicity, require long-term administration and cause serious side-effects, leading to the emergence of resistant and relapse cases. Therefore, there is an urgent need for the discovery of novel antileishmanial molecules and the development of new treatment regimens. The drug used for chemotherapy of B-cell malignancies, Ibrutinib, an inhibitor of Bruton's Tyrosine Kinase (BTK), can offer a new therapeutic perspective due to the functions of BTK on intracellular signaling mechanism of macrophages, which are the primary resident cell for Leishmania. Hence, the study aimed to evaluate ibrutinib as a potential anti-Leishmanial drug. METHOD In this study, we evaluated the antileishmanial effect of Ibrutinib by in vitro L. infantum infection model using macrophages, with cell viability assay, parasite rescue assay, real-time qPCR. RESULTS We showed that Ibrutinib was significantly more effective than the Glucantime against L. infantum. In addition, our data revealed that Ibrutinib inhibited parasite growth and load without impairing macrophage viability. CONCLUSIONS Consequently, due to its efficacy and safety, Ibrutinib may be a promising candidate for the treatment of VL caused by L. infantum as a host-targeted drug.
Collapse
Affiliation(s)
- Ufuk Mert
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey.,Atatürk Health Care Vocational School, Ege University, Izmir, Turkey
| | - Can Müftüoğlu
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Sevgi Erdem
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Aygül Sadıqova
- Infectious Disease Division, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Seray Toz
- Department of Parasitology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Yusuf Ozbel
- Department of Parasitology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Ayse Caner
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey. .,Department of Parasitology, Faculty of Medicine, Ege University, Izmir, Turkey. .,Cancer Research Center, Ege University, Izmir, Turkey.
| |
Collapse
|
46
|
Li J, Jia X, Qiu J, Wang M, Chen J, Jing M, Xu Y, Zheng X, Dai H. Brønsted Acid-Catalyzed Synthesis of 1,2,5-Trisubstituted Imidazoles via a Multicomponent Reaction of Vinyl Azides with Aromatic Aldehydes and Aromatic Amines. J Org Chem 2022; 87:13945-13954. [PMID: 36223536 DOI: 10.1021/acs.joc.2c01624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A facile and efficient approach to the synthesis of 1,2,5-trisubstituted imidazoles is developed via a multicomponent reaction under metal-free catalysis. Under Brønsted acid catalysis, the desired products can be obtained from readily available vinyl azides, aromatic aldehydes, and aromatic amines without generating any toxic waste. The convenient operations and high functional group compatibility indicate that this approach offers an attractive alternative method for the synthesis of imidazole derivatives.
Collapse
Affiliation(s)
- Jiuling Li
- Henan Engineering Research Center of Funiu Mountain's Medical Resources Utilization and Molecular Medicine, School of Medical Sciences, Pingdingshan University, Pingdingshan 467000, People's Republic of China
| | - Xinyu Jia
- Henan Engineering Research Center of Funiu Mountain's Medical Resources Utilization and Molecular Medicine, School of Medical Sciences, Pingdingshan University, Pingdingshan 467000, People's Republic of China
| | - Ju Qiu
- Henan Engineering Research Center of Funiu Mountain's Medical Resources Utilization and Molecular Medicine, School of Medical Sciences, Pingdingshan University, Pingdingshan 467000, People's Republic of China
| | - Min Wang
- Henan Engineering Research Center of Funiu Mountain's Medical Resources Utilization and Molecular Medicine, School of Medical Sciences, Pingdingshan University, Pingdingshan 467000, People's Republic of China
| | - Juan Chen
- Henan Engineering Research Center of Funiu Mountain's Medical Resources Utilization and Molecular Medicine, School of Medical Sciences, Pingdingshan University, Pingdingshan 467000, People's Republic of China
| | - Minghui Jing
- Henan Engineering Research Center of Funiu Mountain's Medical Resources Utilization and Molecular Medicine, School of Medical Sciences, Pingdingshan University, Pingdingshan 467000, People's Republic of China
| | - Yifu Xu
- Henan Engineering Research Center of Funiu Mountain's Medical Resources Utilization and Molecular Medicine, School of Medical Sciences, Pingdingshan University, Pingdingshan 467000, People's Republic of China
| | - Xinhua Zheng
- Henan Engineering Research Center of Funiu Mountain's Medical Resources Utilization and Molecular Medicine, School of Medical Sciences, Pingdingshan University, Pingdingshan 467000, People's Republic of China
| | - Hongmei Dai
- Henan Engineering Research Center of Funiu Mountain's Medical Resources Utilization and Molecular Medicine, School of Medical Sciences, Pingdingshan University, Pingdingshan 467000, People's Republic of China
| |
Collapse
|
47
|
Ma L, Chen X, Zhu S, Chen W, Ma Q, Fan W, Zhang J, Guo L. New β-carboline derivatives containing imidazolium as potential VEGFR2 inhibitors: synthesis, X-ray structure, antiproliferative evaluations, and molecular modeling. RSC Med Chem 2022; 13:1064-1076. [PMID: 36324492 PMCID: PMC9491354 DOI: 10.1039/d2md00065b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/12/2022] [Indexed: 09/19/2023] Open
Abstract
A series of new β-carboline derivatives containing an imidazolium moiety were designed and synthesized via the reaction of β-carboline-1-carboxaldehydes, acetyl chloride, primary amine, and formaldehyde. The antitumor activity of the synthesized compounds was examined against lung carcinoma (A549), gastric carcinoma (BGC-823), murine colon carcinoma (CT-26), liver carcinoma (Bel-7402) and breast carcinoma (MCF-7) cells. The results indicated that most compounds exhibited significant antiproliferative activity, in some cases greater than that of cisplatin, and compound 3z was found to be the most potent antiproliferative agent against A549, BGC823, CT-26, Bel-7402 and MCF-7 cell lines with an IC50 value of 2.7 ± 0.4, 2.7 ± 0.6, 2.4 ± 0.2, 3.2 ± 0.2, and 5.6 ± 0.3 μM, respectively. Combined with favorable in vitro potency, the antitumor efficacies of the selected compounds in mice were also evaluated. Compound 3z exhibited potent antitumor activity with a tumor inhibition rate of 48.6% in sarcoma 180 models. Preliminary investigations on the mechanisms of action revealed that compound 3z could dramatically inhibit EA.hy926 cell tube formation in a dose-dependent manner. Further investigation of the preliminary mechanism of action demonstrated that compound 3z had obvious angiogenesis inhibitory effects in the chicken chorioallantoic membrane (CAM) assay. The results of the docking study showed a good fitting of the new compounds 3o and 3z to the active site of VEGFR-2 with a docking score energy of -11.31 kcal per mole and -11.26 kcal per mole, respectively.
Collapse
Affiliation(s)
- Ling Ma
- School of Chemistry and Chemical Engineering/Key Laboratory for Green Processing of Chemical Engineering of XinJiang Bingtuan, Shihezi University Shihezi China
| | - Xiaofei Chen
- School of Chemistry and Chemical Engineering/Key Laboratory for Green Processing of Chemical Engineering of XinJiang Bingtuan, Shihezi University Shihezi China
| | - Siyu Zhu
- School of Chemistry and Chemical Engineering/Key Laboratory for Green Processing of Chemical Engineering of XinJiang Bingtuan, Shihezi University Shihezi China
| | - Wei Chen
- XinJiang Huashidan Pharmaceutical Research Co. Ltd. Urumqi China
| | - Qin Ma
- XinJiang Huashidan Pharmaceutical Research Co. Ltd. Urumqi China
| | - Wenxi Fan
- XinJiang Huashidan Pharmaceutical Research Co. Ltd. Urumqi China
| | - Jie Zhang
- School of Chemistry and Chemical Engineering/Key Laboratory for Green Processing of Chemical Engineering of XinJiang Bingtuan, Shihezi University Shihezi China
| | - Liang Guo
- School of Chemistry and Chemical Engineering/Key Laboratory for Green Processing of Chemical Engineering of XinJiang Bingtuan, Shihezi University Shihezi China
| |
Collapse
|
48
|
Singh M, Vaishali, Jamra R, Deepika, Kumar S, Singh V. Iodine‐Catalysed Synthesis of β‐Carboline Tethered α‐Amino Amidines Through Ugi‐Type Multicomponent Reaction. ChemistrySelect 2022. [DOI: 10.1002/slct.202202392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Manpreet Singh
- Department of Chemistry Baba Farid Group of Institutions Bathinda Punjab 151001 India
| | - Vaishali
- Department of Chemistry Dr B. R. Ambedkar National Institute of Technology (NIT) Jalandhar Punjab India 144011
| | - Rahul Jamra
- Department of Chemistry Dr B. R. Ambedkar National Institute of Technology (NIT) Jalandhar Punjab India 144011
- Department of Chemistry Central University of Punjab Bathinda Punjab India 151401
| | - Deepika
- Department of Chemistry Dr B. R. Ambedkar National Institute of Technology (NIT) Jalandhar Punjab India 144011
| | - Sunit Kumar
- Department of Chemistry Dr B. R. Ambedkar National Institute of Technology (NIT) Jalandhar Punjab India 144011
| | - Virender Singh
- Department of Chemistry Dr B. R. Ambedkar National Institute of Technology (NIT) Jalandhar Punjab India 144011
- Department of Chemistry Central University of Punjab Bathinda Punjab India 151401
| |
Collapse
|
49
|
Santiago C, Ortega-Tenezaca B, Barbolla I, Fundora-Ortiz B, Arrasate S, Dea-Ayuela MA, González-Díaz H, Sotomayor N, Lete E. Prediction of Antileishmanial Compounds: General Model, Preparation, and Evaluation of 2-Acylpyrrole Derivatives. J Chem Inf Model 2022; 62:3928-3940. [PMID: 35946598 PMCID: PMC9986876 DOI: 10.1021/acs.jcim.2c00731] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In this work, the SOFT.PTML tool has been used to pre-process a ChEMBL dataset of pre-clinical assays of antileishmanial compound candidates. A comparative study of different ML algorithms, such as logistic regression (LOGR), support vector machine (SVM), and random forests (RF), has shown that the IFPTML-LOGR model presents excellent values of specificity and sensitivity (81-98%) in training and validation series. The use of this software has been illustrated with a practical case study focused on a series of 28 derivatives of 2-acylpyrroles 5a,b, obtained through a Pd(II)-catalyzed C-H radical acylation of pyrroles. Their in vitro leishmanicidal activity against visceral (L. donovani) and cutaneous (L. amazonensis) leishmaniasis was evaluated finding that compounds 5bc (IC50 = 30.87 μM, SI > 10.17) and 5bd (IC50 = 16.87 μM, SI > 10.67) were approximately 6-fold more selective than the drug of reference (miltefosine) in in vitro assays against L. amazonensis promastigotes. In addition, most of the compounds showed low cytotoxicity, CC50 > 100 μg/mL in J774 cells. Interestingly, the IFPMTL-LOGR model predicts correctly the relative biological activity of these series of acylpyrroles. A computational high-throughput screening (cHTS) study of 2-acylpyrroles 5a,b has been performed calculating >20,700 activity scores vs a large space of 647 assays involving multiple Leishmania species, cell lines, and potential target proteins. Overall, the study demonstrates that the SOFT.PTML all-in-one strategy is useful to obtain IFPTML models in a friendly interface making the work easier and faster than before. The present work also points to 2-acylpyrroles as new lead compounds worthy of further optimization as antileishmanial hits.
Collapse
Affiliation(s)
- Carlos Santiago
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco / Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain
| | - Bernabé Ortega-Tenezaca
- Department of Computer Science and Information Technologies, University of A Coruña (UDC), 15071, A Coruña, Spain
| | - Iratxe Barbolla
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco / Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.,BIOFISIKA. Basque Center for Biophysics CSIC-UPV/EHU, 48940, Bilbao, Spain
| | - Brenda Fundora-Ortiz
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco / Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain
| | - Sonia Arrasate
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco / Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain
| | - María Auxiliadora Dea-Ayuela
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Universidad CEU Cardenal Herrera, 46115 Alfara del Patriarca, Valencia, Spain
| | - Humberto González-Díaz
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco / Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain.,BIOFISIKA. Basque Center for Biophysics CSIC-UPV/EHU, 48940, Bilbao, Spain.,IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| | - Nuria Sotomayor
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco / Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain
| | - Esther Lete
- Departamento de Química Orgánica e Inorgánica, Facultad de Ciencia y Tecnología, Universidad del País Vasco / Euskal Herriko Unibertsitatea UPV/EHU, Apdo. 644, 48080 Bilbao, Spain
| |
Collapse
|
50
|
Verboni M, Olivieri D, Lucarini S. A recent update on new synthetic chiral compounds with antileishmanial activity. Chirality 2022; 34:1279-1297. [PMID: 35947400 PMCID: PMC9543214 DOI: 10.1002/chir.23494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/21/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022]
Abstract
Parasitic diseases, including malaria, leishmaniasis, and trypanosomiasis, affect billions of people and are responsible for almost 500,000 deaths/year. In particular, leishmaniasis, a neglected tropical disease, is considered a global public health problem because current drugs have several drawbacks including to toxicity, high cost, and drug resistance, which result in a lack of effective and readily available therapies. Therefore, the synthesis of new, safe, and effective molecules still requires the attention of the scientific community. Moreover, it is well known that chirality plays a crucial role in the antiparasitic activity of molecules, driving the design of their synthesis. Therefore, in this review we report a recent update on new chiral compounds with promising antileishmanial activity, focusing on synthetic approaches. Where reported, in most cases the enantiopure compound has shown better potency against the protozoa than its enantiomer or corresponding racemic mixture.
Collapse
Affiliation(s)
- Michele Verboni
- Department of Biomolecular Science, University of Urbino Carlo Bo, Urbino, Italy
| | - Diego Olivieri
- Department of Biomolecular Science, University of Urbino Carlo Bo, Urbino, Italy
| | - Simone Lucarini
- Department of Biomolecular Science, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|