1
|
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
|
2
|
Vasanthan RJ, Pradhan S, Thangamuthu MD. Emerging Aspects of Triazole in Organic Synthesis: Exploring its Potential as a Gelator. Curr Org Synth 2024; 21:456-512. [PMID: 36221871 DOI: 10.2174/1570179420666221010094531] [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: 05/19/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 11/22/2022]
Abstract
Cu(I)-catalyzed azide-alkyne 1,3-dipolar cycloaddition (CuAAC) - commonly known as the "click reaction" - serves as the most effective and highly reliable tool for facile construction of simple to complex designs at the molecular level. It relates to the formation of carbon heteroatomic systems by joining or clicking small molecular pieces together with the help of various organic reactions such as cycloaddition, conjugate addition, ring-opening, etc. Such dynamic strategy results in the generation of triazole and its derivatives from azides and alkynes with three nitrogen atoms in the five-membered aromatic azole ring that often forms gel-assembled structures having gelating properties. These scaffolds have led to prominent applications in designing advanced soft materials, 3D printing, ion sensing, drug delivery, photonics, separation, and purification. In this review, we mainly emphasize the different mechanistic aspects of triazole formation, which includes the synthesis of sugar-based and non-sugar-based triazoles, and their gel applications reported in the literature for the past ten years, as well as the upcoming scope in different branches of applied sciences.
Collapse
Affiliation(s)
- Rabecca Jenifer Vasanthan
- Department of Chemistry, School of Basic and Applied Sciences, Central University of Tamil Nadu (CUTN), Thiruvarur, 610 005, India
| | - Sheersha Pradhan
- Department of Chemistry, School of Basic and Applied Sciences, Central University of Tamil Nadu (CUTN), Thiruvarur, 610 005, India
| | - Mohan Das Thangamuthu
- Department of Chemistry, School of Basic and Applied Sciences, Central University of Tamil Nadu (CUTN), Thiruvarur, 610 005, India
| |
Collapse
|
3
|
Stereoselective Synthesis and Application of Gibberellic Acid-Derived Aminodiols. Int J Mol Sci 2022; 23:ijms231810366. [PMID: 36142293 PMCID: PMC9499365 DOI: 10.3390/ijms231810366] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/27/2022] [Accepted: 09/05/2022] [Indexed: 11/28/2022] Open
Abstract
A series of gibberellic acid-based aminodiols was designed and synthesized from commercially available gibberellic acid. Exposure of gibberellic acid to hydrochloric acid under reflux conditions resulted in aromatization followed by rearrangement to form allo-gibberic acid. The key intermediate, ethyl allo-gibberate, was prepared according to literature methods. Epoxidation of key intermediate and subsequent ring-opening of the corresponding epoxide with different nucleophiles resulted in N-substituted aminodiols. The regioselective ring closure of N-benzyl-substituted aminodiol with formaldehyde was also investigated. All aminodiol derivatives were well characterized using modern spectroscopic techniques and evaluated for their antiproliferative activity against a panel of human cancer cell lines. In addition, structure–activity relationships were examined by assessing substituent effects on the aminodiol systems. The results indicated that aminodiols containing aromatic rings on their nitrogen substituents displayed significant cytotoxic effects. Among these agents, N-naphthylmethyl-substituted aminodiols were found to be the most potent candidates in this series. One of these molecules exhibited a modest cancer selectivity determined by non-cancerous fibroblast cells. A docking study was also made to exploit the observed results.
Collapse
|
4
|
Raut DG, Bhosale RB, Lawand AS, Hublikar MG, Kadu VD, Patil SB, Choudhari PB. Syntheses, Molecular Docking and Biological Evaluation of 2-(2- hydrazinyl)thiazoles as Potential Antioxidant, Anti-Inflammatory and Significant Anticancer Agents. RECENT ADVANCES IN INFLAMMATION & ALLERGY DRUG DISCOVERY 2022; 16:96-106. [PMID: 36056853 DOI: 10.2174/2772270816666220902094019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND Recently, researchers have worked on the development of new methods for the synthesis of bioactive heterocycles using polyethylene glycol as a green solvent. In this context, we report the synthesized 2-(2-hydrazinyl) thiazoles for their in vitro antioxidant, in vitro anti-inflammatory and in vitro anti-cancer activities. OBJECTIVE The objective of the study was to develop novel antioxidant, anti-inflammatory and anti-cancer drugs. METHODS At the outset, the condensation of substituted acetophenones 1, thiosemicarbazide 2, and α-haloketones 3 was carried out using PEG-400 (20 mL) in the presence of 5 mol% glacial acetic acid to afford thiosemicarbazones intermediate. Furthermore, these thiosemicarbazones were reacted with α-haloketones 3 to obtain appropriate 2-(2-hydrazinyl) thiazoles. The synthesized compounds were in vitro tested for their antioxidant, anti-inflammatory, and anti-cancer activity. RESULTS In vitro evaluation report showed that nearly all molecules possessed potential antioxidant activity against 2,2-Diphenyl-1-picrylhydrazyl (DPPH), nitric oxide (NO), superoxide radical (SOR) and hydrogen peroxide (H2O2) radical scavenging activity. Most 2-(2-hydrazinyl) thiazoles derivatives have shown potential anti-inflammatory activity as compared to diclofenac sodium as a reference standard. 2-(2-Hydrazinyl) thiazoles derivatives showed significant anticancer activity for human leukemia cell line K-562 compared to adriamycin as a reference standard. CONCLUSION All tested compounds showed potential 2,2-Diphenyl-1-picrylhydrazyl (DPPH) and nitric oxide (NO) radical scavenging activity. Among the tested series, 4b, 4d and 4e exhibited good hydrogen peroxide and 4b, 4e, 4f and 4g showed excellent superoxide radical scavenging activity. In addition, the 4b, 4e and 4g compounds revealed potent in vitro anti-inflammatory activity against standard diclofenac sodium drug. 2-(2-Hydrazinyl) thiazole derivatives, such as 4c and 4d, showed significant anticancer activity against human leukemia cell line K-562. Thus, these molecules provide an interesting template for the design and development of new antioxidant, anti-inflammatory, and anti-cancer agents.
Collapse
Affiliation(s)
- Dattatraya G Raut
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Punyashlok Ahilyadevi Holkar Solapur University, Solapur - 413255 Maharashtra, India
| | - Raghunath B Bhosale
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Punyashlok Ahilyadevi Holkar Solapur University, Solapur - 413255 Maharashtra, India
| | - Anjana S Lawand
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Punyashlok Ahilyadevi Holkar Solapur University, Solapur - 413255 Maharashtra, India
| | - Mahesh G Hublikar
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Punyashlok Ahilyadevi Holkar Solapur University, Solapur - 413255 Maharashtra, India
| | - Vikas D Kadu
- Organic Chemistry Research Laboratory, School of Chemical Sciences, Punyashlok Ahilyadevi Holkar Solapur University, Solapur - 413255 Maharashtra, India
| | - Sandeep B Patil
- Department of Pharmacology, Dr. Shivajirao Kadam College of Pharmacy Kasbe Digraj, Sangli, Maharashtra, India
| | - Prafulla B Choudhari
- Computational Chemistry Research Lab, Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, 416013 Maharashtra, India
| |
Collapse
|
5
|
Mousavi A, Foroumadi P, Emamgholipour Z, Mäser P, Kaiser M, Foroumadi A. 2-(Nitroaryl)-5-Substituted-1,3,4-Thiadiazole Derivatives with Antiprotozoal Activities: In Vitro and In Vivo Study. Molecules 2022; 27:molecules27175559. [PMID: 36080325 PMCID: PMC9457997 DOI: 10.3390/molecules27175559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Nitro-containing compounds are a well-known class of anti-infective agents, especially in the field of anti-parasitic drug discovery. HAT or sleeping sickness is a neglected tropical disease caused by a protozoan parasite, Trypanosoma brucei. Following the approval of fexinidazole as the first oral treatment for both stages of T. b. gambiense HAT, there is an increased interest in developing new nitro-containing compounds against parasitic diseases. In our previous projects, we synthesized several megazole derivatives that presented high activity against Leishmania major promastigotes. Here, we screened and evaluated their trypanocidal activity. Most of the compounds showed submicromolar IC50 against the BSF form of T. b. rhodesiense (STIB 900). To the best of our knowledge, compound 18c is one of the most potent nitro-containing agents reported against HAT in vitro. Compound 18g revealed an acceptable cure rate in the acute mouse model of HAT, accompanied with noteworthy in vitro activity against T. brucei, T. cruzi, and L. donovani. Taken together, these results suggest that these compounds are promising candidates to evaluate their pharmacokinetic and biological profiles in the future.
Collapse
Affiliation(s)
- Alireza Mousavi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Parham Foroumadi
- Department of Medicinal Chemistry, School of Pharmacy, International Campus, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Zahra Emamgholipour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland
- Faculty of Science, University of Basel, 4002 Basel, Switzerland
- Correspondence: (M.K.); (A.F.)
| | - Alireza Foroumadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
- Correspondence: (M.K.); (A.F.)
| |
Collapse
|
6
|
Shahidul Islam M, Al‐Majid AM, Azam M, Prakash Verma V, Barakat A, Haukka M, Domingo LR, Elgazar AA, Mira A, Badria FA. Synthesis of Spirooxindole Analogs Tethered Pyrazole Scaffold as Acetylcholinesterase Inhibitors. ChemistrySelect 2021. [DOI: 10.1002/slct.202103255] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Mohammad Shahidul Islam
- Department of Chemistry College of Science King Saud University P.O. Box 2455 Riyadh 11451 Saudi Arabia
- Department of Chemistry Banasthali Vidyapith Banasthali- 304022 Rajasthan India
| | | | - Mohammad Azam
- Department of Chemistry College of Science King Saud University P.O. Box 2455 Riyadh 11451 Saudi Arabia
| | - Ved Prakash Verma
- Department of Chemistry Banasthali Vidyapith Banasthali- 304022 Rajasthan India
| | - Assem Barakat
- Department of Chemistry College of Science King Saud University P.O. Box 2455 Riyadh 11451 Saudi Arabia
- Department of Chemistry, Faculty of Science Alexandria University, P.O. Box 426, Ibrahimia Alexandria 21321 Egypt
| | - Matti Haukka
- Department of Chemistry University of Jyväskylä, P.O. Box 35 FI-40014 Jyväskylä Finland
| | - Luis R. Domingo
- Department of Organic Chemistry University of Valencia, Dr. Moliner 50 46100 Burjassot, Valencia Spain
| | - Abdullah A. Elgazar
- Department of Pharmacognosy, Faculty of Pharmacy Kafrelsheikh University Kafrelsheikh 33516 Egypt
| | - Amira Mira
- Department of Pharmacognosy, Faculty of Pharmacy Mansoura University Mansoura 35516 Egypt
| | - Farid A. Badria
- Department of Pharmacognosy, Faculty of Pharmacy Mansoura University Mansoura 35516 Egypt
| |
Collapse
|
7
|
Islam M, Al-Majid AM, Azam M, Verma VP, Barakat A, Haukka M, Elgazar AA, Mira A, Badria FA. Construction of Spirooxindole Analogues Engrafted with Indole and Pyrazole Scaffolds as Acetylcholinesterase Inhibitors. ACS OMEGA 2021; 6:31539-31556. [PMID: 34869980 PMCID: PMC8637602 DOI: 10.1021/acsomega.1c03978] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/01/2021] [Indexed: 05/12/2023]
Abstract
Twenty-five new hits of spirooxindole analogs 8a-y engrafted with indole and pyrazole scaffolds were designed and constructed via a [3+2]cycloaddition (32CA) reaction starting from three components: new chalcone-based indole and pyrazole scaffolds 5a-d, substituted isatins 6a-c, and secondary amines 7a-d. The potency of the compounds were assessed in modulating cholinesterase (AChE) activity using Ellman's method. Compounds 8i and 8y showed the strongest acetylcholine esterase inhibition (AChEI) with IC50 values of 24.1 and 27.8 μM, respectively. Molecular docking was used to study their interaction with the active site of hAChE.
Collapse
Affiliation(s)
- Mohammad
Shahidul Islam
- Department
of Chemistry, College of Science, King Saud
University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
- Department
of Chemistry, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Abdullah Mohammed Al-Majid
- Department
of Chemistry, College of Science, King Saud
University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Mohammad Azam
- Department
of Chemistry, College of Science, King Saud
University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Ved Prakash Verma
- Department
of Chemistry, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Assem Barakat
- Department
of Chemistry, College of Science, King Saud
University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
- Department
of Chemistry, Faculty of Science, Alexandria
University, P.O. Box 426, Ibrahimia, Alexandria 21321, Egypt
| | - Matti Haukka
- Department
of Chemistry, University of Jyväskylä, P.O. Box 35, FI-40014 Jyväskylä, Finland
| | - Abdullah A. Elgazar
- Department
of Pharmacognosy, Faculty of Pharmacy, Kafrelsheikh
University, Kafrelsheikh 33516, Egypt
| | - Amira Mira
- Department
of Pharmacognosy, Faculty of Pharmacy, Mansoura
University, Mansoura 35516, Egypt
| | - Farid A. Badria
- Department
of Pharmacognosy, Faculty of Pharmacy, Mansoura
University, Mansoura 35516, Egypt
| |
Collapse
|
8
|
Cyr P, Joseph‐Valcin E, Boissarie P, Simoneau B, Marinier A. Copper‐Catalyzed
N
1
Coupling of 3‐Aminoindazoles and Related Aminoazoles with Aryl Bromides. European J Org Chem 2021. [DOI: 10.1002/ejoc.202101245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Patrick Cyr
- Medicinal Chemistry Institute of Research in Immunology and Cancer Université de Montréal Montreal H3C 3J7 Qc Canada
| | - Eve‐Marline Joseph‐Valcin
- Medicinal Chemistry Institute of Research in Immunology and Cancer Université de Montréal Montreal H3C 3J7 Qc Canada
| | - Patrick Boissarie
- Medicinal Chemistry Institute of Research in Immunology and Cancer Université de Montréal Montreal H3C 3J7 Qc Canada
| | - Bruno Simoneau
- Medicinal Chemistry Institute of Research in Immunology and Cancer Université de Montréal Montreal H3C 3J7 Qc Canada
| | - Anne Marinier
- Medicinal Chemistry Institute of Research in Immunology and Cancer Université de Montréal Montreal H3C 3J7 Qc Canada
- Département de chimie Faculté des Arts et Sciences Université de Montréal Montreal H2V 0B3 Qc Canada
- Département de Pharmacologie Faculté de Médecine Université de Montréal Montreal H3C 3J7 Qc Canada
| |
Collapse
|
9
|
Chen Q, Wu C, Zhu J, Li E, Xu Z. Therapeutic potential of indole derivatives as anti-HIV agents: A mini-review. Curr Top Med Chem 2021; 22:993-1008. [PMID: 34636313 DOI: 10.2174/1568026621666211012111901] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/22/2021] [Accepted: 08/28/2021] [Indexed: 11/22/2022]
Abstract
Acquired immunodeficiency syndrome (AIDS), caused by human immunodeficiency virus (HIV), is one of the leading causes of human deaths. The advent of different anti-HIV drugs over different disease progress has made AIDS/HIV from a deadly infection to chronic and manageable disease. However, the development of multidrug-resistant viruses, together with the severe side effects of anti-HIV agents, compromised their efficacy and limited the treatment options. Indoles, the most common frameworks in the bioactive molecules, represent attractive scaffolds for the design and development of novel drugs. Indole derivatives are potential inhibitors of HIV enzymes such as reverse transcriptase, integrase and protease, and some indole-based agents like Delavirdine have already been applied in clinics or under clinical evaluations for the treatment of AIDS/HIV, revealing that indole moiety is a useful template for the development of anti-HIV agents. This review focuses on the recent advancement of indole derivatives including indole alkaloids, hybrids, and dimers with anti-HIV potential, covering articles published between 2010 and 2020. The chemical structures, structure-activity relationship and mechanisms of action are also discussed.
Collapse
Affiliation(s)
- Qingtai Chen
- School of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian, 463000. China
| | - Chongchong Wu
- Department of Chemical and Petroleum Engineering, University of Calgary, T2N 1N4 Calgary, Alberta. Canada
| | - Jinjin Zhu
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian, 463000. China
| | - Enzhong Li
- School of Biological and Food Processing Engineering, Huanghuai University, Zhumadian, 463000. China
| | - Zhi Xu
- School of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian, 463000. China
| |
Collapse
|
10
|
Prasher P, Sharma M, Zacconi F, Gupta G, Aljabali AA, Mishra V, Tambuwala MM, Kapoor DN, Negi P, Andreoli Pinto TDJ, Singh I, Chellappan DK, Dua K. Synthesis and Anticancer Properties of ‘Azole’ Based Chemotherapeutics as Emerging Chemical Moieties: A Comprehensive Review. CURR ORG CHEM 2021. [DOI: 10.2174/1385272824999200820152501] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Azole frameworks serve as privileged scaffolds in the contemporary drug design
paradigm owing to their unique physicochemical profile that promotes the development
of highly selective, physiological benevolent chemotherapeutics. Several azole nuclei
function as bioisostere in medicinal chemistry and prompt the development of tailored
therapeutics for targeting the desired biological entities. Besides, the azole scaffold forms
an integral part in the advanced drug designing methodologies, such as target template insitu
drug synthesis, that assists in rapid identification of the hit molecules form a diverse
pool of leads; and direct biomolecule-drug conjugation, along with bioorthogonal strategies
that ensure localization, and superior target specificity of the directed therapeutic.
Lastly, the structural diversity of azole framework and high yielding click synthetic methods
provide a comprehensive Structure-Activity Relationship analysis for design optimization of the potential
drug molecules by fine-tuning the placement of different substituents critical for the activity. This review provides
a comprehensive analysis of the synthesis and anticancer potential of azole based chemotherapeutics.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun 248007, India
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Arcadia Grant, Dehradun 248007, India
| | - Flavia Zacconi
- Departamento de Quimica Organica, Facultad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile, Av. Vicuna Mackenna 4860, Macul, Santiago 7820436, Chile
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302 017, Jaipur, India
| | - Alaa A.A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, County Londonderry, Northern Ireland BT52 1SA, United Kingdom
| | - Deepak N. Kapoor
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Post box no. 9, Solan, Himachal Pradesh 173 229, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Post box no. 9, Solan, Himachal Pradesh 173 229, India
| | - Terezinha de Jesus Andreoli Pinto
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of São Paulo, Professor Lineu Prestes Street, São Paulo 05508-000, Brazil
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Dinesh K. Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
11
|
Triazole-containing hybrids with anti- Mycobacterium tuberculosis potential - Part I: 1,2,3-Triazole. Future Med Chem 2021; 13:643-662. [PMID: 33619989 DOI: 10.4155/fmc-2020-0301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Tuberculosis regimens currently applied in clinical practice require months of multidrug therapy, which imposes a major challenge of patient compliance and drug resistance development. Moreover, because of the increasing emergence of hard-to-treat tuberculosis, this disease continues to be a significant threat to the human population. 1,2,3-triazole as a privileged structure has been widely used as an effective template for drug discovery, and 1,2,3-triazole-containing hybrids that can simultaneously act on dual or multiple targets in Mycobacterium tuberculosis have the potential to circumvent drug resistance, enhance efficacy, reduce side effects and improve pharmacokinetic as well as pharmacodynamic profiles. Thus, 1,2,3-triazole-containing hybrids are useful scaffolds for the development of antitubercular agents. This review aims to highlight recent advances of 1,2,3-triazole-containing hybrids with potential activity against various forms of M. tuberculosis, covering articles published between 2015 and 2020. The structure-activity relationship and the mechanism of action are also discussed to facilitate further rational design of more effective drug candidates.
Collapse
|
12
|
1,2,3-Triazole-containing hybrids with potential antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA). Eur J Med Chem 2020; 206:112686. [PMID: 32795773 DOI: 10.1016/j.ejmech.2020.112686] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 01/10/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA), as a classic reason for genuine skin and flimsy tissues diseases, is a worldwide general wellbeing risk and has already tormented humanity for a long history, creating a critical need for the development of new classes of antibacterials. 1,2,3-Triazole moiety, readily interact with diverse enzymes and receptors in organisms through weak bond interaction, is among the most common frameworks present in the bioactive molecules. 1,2,3-Triazole derivatives, especially 1,2,3-triazole-containing hybrids, possess broad-spectrum activity against a panel of clinically important bacteria including drug-resistant pathogens, so rational design of 1,2,3-triazole derivatives may open a door for the opportunities on the development of novel anti-MRSA agents. This review is an endeavour to highlight the current scenario of 1,2,3-triazole-containing hybrids with potential anti-MRSA activity, covering articles published between 2010 and 2020.
Collapse
|