1
|
Dasmahapatra U, Maiti B, Chanda K. A microwave assisted tandem synthesis of quinazolinones using ionic liquid supported copper(II) catalyst with mechanistic insights. Org Biomol Chem 2024; 22:8459-8471. [PMID: 39320933 DOI: 10.1039/d4ob01261e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Quinazolinone is a preferred structural motif with notable pharmacological activity that is present in a wide range of naturally occurring compounds. A microwave assisted tandem cyclooxidative method has been developed to afford quinazolinones via a recyclable ionic liquid supported copper catalyst. This sustainable method exhibits operational simplicity through a rapid, clean, and energy-efficient route and a variety of 2-substituted quinazolinones are obtained in excellent yields. In addition, this innovative approach enables us to develop a library of nitriles in an environment-friendly synthetic protocol. Moreover, the catalyst can be recycled and reused up to three consecutive cycles without any significant loss of catalytic activity. Further organic transformation of the synthesized quinazolinones was carried out to afford reported as well as novel bioactive heterocyclic compounds.
Collapse
Affiliation(s)
- Upala Dasmahapatra
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore- 632014, India
| | - Barnali Maiti
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore- 632014, India
| | - Kaushik Chanda
- Department of Chemistry, Rabindranath Tagore University, Hojai, Assam-782435, India.
| |
Collapse
|
2
|
Petzold Pauli F, de Oliveira Lima Filho E, de Almeida Ribeiro Oliveira G, Morais Lião L, Rangel Campos V, da Silva Magalhães Forezi L, Francisco Ferreira V, de Carvalho da Silva F. Divergent Regioselective Synthesis of Functionalized 1,2,3-1H-Triazoles from Nitriles and Arylazides Under Metal-Free and/or Solvent-Free Conditions. Chem Asian J 2024:e202400845. [PMID: 39352621 DOI: 10.1002/asia.202400845] [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: 07/16/2024] [Revised: 09/18/2024] [Indexed: 11/09/2024]
Abstract
Highly selective and divergent syntheses, which are crucial in both organic synthesis and medicinal chemistry, involve significant advancements in compound accessibility. By modifying α-cyano esters into α-cyano ketones, the synthesis pathway broadens to include a diverse range of 4-CN, 5-amino, and 5-arylamino derivatives of 1,2,3-triazoles, which are achieved notably through the Dimroth rearrangement. This versatility extends further with the potential for a triple cascade reaction, leading to the production of carboximidamide compounds, which are facilitated by the Cornforth rearrangement. Advancements in compound accessibility not only expand the repertoire of synthesized molecules but also open new avenues for potential pharmacological agents. Building on these findings, we have developed an innovative and efficient method for the divergent synthesis of functionalized 1,2,3-triazoles. This method strategically utilizes α-cyanocarbonyls and arylazides by harnessing their reactivity and compatibility to orchestrate a variety of molecular transformations. By optimizing these substrates, our goal is to simplify synthetic routes, improve product yields, and accelerate the discovery and development of new chemical entities with promising biological activities.
Collapse
Affiliation(s)
- Fernanda Petzold Pauli
- Universidade do Estado do Rio de Janeiro, Instituto de Química, Departamento de Química Orgânica, Rio de Janeiro-RJ, Brazil
| | | | | | | | - Vinicius Rangel Campos
- Universidade Federal Fluminense, Instituto de Química, Departamento de Química Orgânica, Niterói-RJ, Brazil
| | | | - Vitor Francisco Ferreira
- Universidade Federal Fluminense, Faculdade de Farmácia, Departamento de Tecnologia Farmacêutica, Niterói-RJ, Brazil
| | | |
Collapse
|
3
|
Nyame P, Togami A, Yoshida T, Masunaga T, Begum MM, Terasawa H, Monde N, Tahara Y, Tanaka R, Tanaka Y, Appiah-Kubi J, Amesimeku WAO, Hossain MJ, Otsuka M, Yoshimura K, Ikeda T, Sawa T, Satou Y, Fujita M, Maeda Y, Tateishi H, Monde K. A heterocyclic compound inhibits viral release by inducing cell surface BST2/Tetherin/CD317/HM1.24. J Biol Chem 2024; 300:107701. [PMID: 39173946 PMCID: PMC11419809 DOI: 10.1016/j.jbc.2024.107701] [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: 05/31/2024] [Revised: 08/01/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024] Open
Abstract
The introduction of combined antiretroviral therapy (cART) has greatly improved the quality of life of human immunodeficiency virus type 1 (HIV-1)-infected individuals. Nonetheless, the ever-present desire to seek out a full remedy for HIV-1 infections makes the discovery of novel antiviral medication compelling. Owing to this, a new late-stage inhibitor, Lenacapavir/Sunlenca, an HIV multi-phase suppressor, was clinically authorized in 2022. Besides unveiling cutting-edge antivirals inhibiting late-stage proteins or processes, newer therapeutics targeting host restriction factors hold promise for the curative care of HIV-1 infections. Notwithstanding, bone marrow stromal antigen 2 (BST2)/Tetherin/CD317/HM1.24, which entraps progeny virions is an appealing HIV-1 therapeutic candidate. In this study, a novel drug screening system was established, using the Jurkat/Vpr-HiBiT T cells, to identify drugs that could obstruct HIV-1 release; the candidate compounds were selected from the Ono Pharmaceutical compound library. Jurkat T cells expressing Vpr-HiBiT were infected with NL4-3, and the amount of virus release was quantified indirectly by the amount of Vpr-HiBiT incorporated into the progeny virions. Subsequently, the candidate compounds that suppressed viral release were used to synthesize the heterocyclic compound, HT-7, which reduces HIV-1 release with less cellular toxicity. Notably, HT-7 increased cell surface BST2 coupled with HIV-1 release reduction in Jurkat cells but not Jurkat/KO-BST2 cells. Seemingly, HT-7 impeded simian immunodeficiency virus (SIV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) release. Concisely, these results suggest that the reduction in viral release, following HT-7 treatment, resulted from the modulation of cell surface expression of BST2 by HT-7.
Collapse
Affiliation(s)
- Perpetual Nyame
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Akihiro Togami
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomofumi Yoshida
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takuya Masunaga
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Mst Monira Begum
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Hiromi Terasawa
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Nami Monde
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yurika Tahara
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Reiko Tanaka
- Laboratory of Hemato-Immunology, Graduate School of Health Sciences, University of the Ryukyus, Okinawa, Japan
| | - Yuetsu Tanaka
- Laboratory of Hemato-Immunology, Graduate School of Health Sciences, University of the Ryukyus, Okinawa, Japan
| | - Joyce Appiah-Kubi
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Md Jakir Hossain
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Masami Otsuka
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Department of Drug Discovery, Science Farm Ltd, Kumamoto, Japan
| | - Kazuhisa Yoshimura
- Department of Microbiology, Tokyo Metropolitan Institute of Public Health, Tokyo, Japan
| | - Terumasa Ikeda
- Division of Molecular Virology and Genetics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yorifumi Satou
- Division of Genomics and Transcriptomics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Mikako Fujita
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yosuke Maeda
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Department of Nursing, Kibi International University, Takahashi, Japan
| | - Hiroshi Tateishi
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Research & Development, Hirata Corporation, Kumamoto, Japan.
| | - Kazuaki Monde
- Department of Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; Collaboration Unit for Infection, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
4
|
Raman APS, Aslam M, Awasthi A, Ansari A, Jain P, Lal K, Bahadur I, Singh P, Kumari K. An updated review on 1,2,3-/1,2,4-triazoles: synthesis and diverse range of biological potential. Mol Divers 2024:10.1007/s11030-024-10858-0. [PMID: 39066993 DOI: 10.1007/s11030-024-10858-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/22/2024] [Indexed: 07/30/2024]
Abstract
The synthesis of triazoles has attracted a lot of interest in the field of organic chemistry because of its versatile chemical characteristics and possible biological uses. This review offers an extensive overview of the different pathways used in the production of triazoles. A detailed analysis of recent research indicates that triazole compounds have a potential range of pharmacological activities, including the ability to inhibit enzymes, and have antibacterial, anticancer, and antifungal activities. The integration of computational and experimental methods provides a thorough understanding of the structure-activity connection, promoting sensible drug design and optimization. By including triazoles as essential components in drug discovery, researchers can further explore and innovate in the synthesis, biological assessment, and computational studies of triazoles as drugs, exploring the potential therapeutic significance of triazoles.
Collapse
Affiliation(s)
- Anirudh Pratap Singh Raman
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, Dhaula Kuan, New Delhi, India
- Department of Chemistry, SRM Institute of Science & Technology, Delhi-NCR Campus, Ghaziabad, Modinagar, India
| | - Mohd Aslam
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, Dhaula Kuan, New Delhi, India
- Department of Chemistry, SRM Institute of Science & Technology, Delhi-NCR Campus, Ghaziabad, Modinagar, India
| | - Amardeep Awasthi
- Department of Chemistry, North western University, Evanston, IL, USA
| | - Anas Ansari
- Department of Chemistry, North western University, Evanston, IL, USA
| | - Pallavi Jain
- Department of Chemistry, SRM Institute of Science & Technology, Delhi-NCR Campus, Ghaziabad, Modinagar, India
| | - Kashmiri Lal
- Department of Chemistry, Guru Jambheshwar of Science and Technology, Hisar, India
| | - Indra Bahadur
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, North-West University, Mafikeng Campus, Mmabatho, 2745, South Africa
| | - Prashant Singh
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, Dhaula Kuan, New Delhi, India.
| | - Kamlesh Kumari
- Department of Zoology, University of Delhi, Delhi, India.
| |
Collapse
|
5
|
Lang S, Fletcher DA, Petit AP, Luise N, Fyfe P, Zuccotto F, Porter D, Hope A, Bellany F, Kerr C, Mackenzie CJ, Wyatt PG, Gray DW. Application of an NMR/Crystallography Fragment Screening Platform for the Assessment and Rapid Discovery of New HIV-CA Binding Fragments. ChemMedChem 2024; 19:e202400025. [PMID: 38581280 DOI: 10.1002/cmdc.202400025] [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: 01/26/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/08/2024]
Abstract
Identification and assessment of novel targets is essential to combat drug resistance in the treatment of HIV/AIDS. HIV Capsid (HIV-CA), the protein playing a major role in both the early and late stages of the viral life cycle, has emerged as an important target. We have applied an NMR fragment screening platform and identified molecules that bind to the N-terminal domain (NTD) of HIV-CA at a site close to the interface with the C-terminal domain (CTD). Using X-ray crystallography, we have been able to obtain crystal structures to identify the binding mode of these compounds. This allowed for rapid progression of the initial, weak binding, fragment starting points to compounds 37 and 38, which have 19F-pKi values of 5.3 and 5.4 respectively.
Collapse
Affiliation(s)
- Stuart Lang
- Cresset Discovery, New Cambridge House, Bassingbourn Road, Litlington, Cambridgeshire, SG80SSS
| | - Daniel A Fletcher
- BioAscent Discovery Ltd, Bo'Ness Road, Newhouse, Lanarkshire, ML1 5UH
| | | | - Nicola Luise
- Alira Health, Av. De Josep Tarradellas, 123, 7th Floor, 08029, Barcelona, Spain
| | - Paul Fyfe
- Drug Discovery Unit, University of Dundee, Dow Street, Dundee, DD1 5EH
| | - Fabio Zuccotto
- Vertex Pharmaceuticals (Europe) Ltd, 86-88, Jubilee Avenue, Milton Park, Abingdon, Oxfordshire, OX14 4RW
| | - David Porter
- Evotec (UK) Ltd, Dorothy Crowfoot Hodgkin Campus, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire, OX14 4RZ
| | - Anthony Hope
- Drug Discovery Unit, University of Dundee, Dow Street, Dundee, DD1 5EH
| | - Fiona Bellany
- Drug Discovery Unit, University of Dundee, Dow Street, Dundee, DD1 5EH
| | - Catrina Kerr
- Drug Discovery Unit, University of Dundee, Dow Street, Dundee, DD1 5EH
| | | | - Paul G Wyatt
- Sitala Bio Ltd, Unit D6, Grain House Mill Court, Great Shelford, Cambridge, CB22 5LD
| | - David W Gray
- Drug Discovery Unit, University of Dundee, Dow Street, Dundee, DD1 5EH
| |
Collapse
|
6
|
Ahmad G, Sohail M, Bilal M, Rasool N, Qamar MU, Ciurea C, Marceanu LG, Misarca C. N-Heterocycles as Promising Antiviral Agents: A Comprehensive Overview. Molecules 2024; 29:2232. [PMID: 38792094 PMCID: PMC11123935 DOI: 10.3390/molecules29102232] [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: 03/07/2024] [Revised: 04/22/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Viruses are a real threat to every organism at any stage of life leading to extensive infections and casualties. N-heterocycles can affect the viral life cycle at many points, including viral entrance into host cells, viral genome replication, and the production of novel viral species. Certain N-heterocycles can also stimulate the host's immune system, producing antiviral cytokines and chemokines that can stop the reproduction of viruses. This review focused on recent five- or six-membered synthetic N-heterocyclic molecules showing antiviral activity through SAR analyses. The review will assist in identifying robust scaffolds that might be utilized to create effective antiviral drugs with either no or few side effects.
Collapse
Affiliation(s)
- Gulraiz Ahmad
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan; (G.A.); (M.S.)
| | - Maria Sohail
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan; (G.A.); (M.S.)
| | - Muhammad Bilal
- School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China;
| | - Nasir Rasool
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan; (G.A.); (M.S.)
| | - Muhammad Usman Qamar
- Institute of Microbiology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan;
- Division of Infectious Diseases, Geneva University Hospitals, 1205 Geneva, Switzerland
- Department of Microbiology and Molecular Medicine, University of Geneva, 1205 Geneva, Switzerland
| | - Codrut Ciurea
- Faculty of Medicine, Transilvania University of Brasov, 500036 Brasov, Romania; (L.G.M.)
| | - Luigi Geo Marceanu
- Faculty of Medicine, Transilvania University of Brasov, 500036 Brasov, Romania; (L.G.M.)
| | - Catalin Misarca
- Faculty of Medicine, Transilvania University of Brasov, 500036 Brasov, Romania; (L.G.M.)
| |
Collapse
|
7
|
Hou X, Yan D, Wu Z, Mao L, Wang H, Guo Y, Yang J. Discovery of Dolutegravir Derivative against Liver Cancer via Inducing Autophagy and DNA Damage. Molecules 2024; 29:1779. [PMID: 38675599 PMCID: PMC11052077 DOI: 10.3390/molecules29081779] [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: 02/22/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
We introduced a terminal alkyne into the core structure of dolutegravir, resulting in the synthesis of 34 novel dolutegravir-1,2,3-triazole compounds through click chemistry. These compounds exhibited remarkable inhibitory activities against two hepatocellular carcinoma cell lines, Huh7 and HepG2. Notably, compounds 5e and 5p demonstrated exceptional efficacy, particularly against Huh7 cells, with IC50 values of 2.64 and 5.42 μM. Additionally, both compounds induced apoptosis in Huh7 cells, suppressed tumor cell clone formation, and elevated reactive oxygen species (ROS) levels, further promoting tumor cell apoptosis. Furthermore, compounds 5e and 5p activated the LC3 signaling pathway, inducing autophagy, and triggered the γ-H2AX signaling pathway, resulting in DNA damage in tumor cells. Compound 5e exhibited low toxicity, highlighting its potential as a promising anti-tumor drug.
Collapse
Affiliation(s)
- Xixi Hou
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China;
| | - Dong Yan
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, 263 Kaiyuan Road, Luoyang 471003, China (Z.W.); (L.M.)
| | - Ziyuan Wu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, 263 Kaiyuan Road, Luoyang 471003, China (Z.W.); (L.M.)
| | - Longfei Mao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, 263 Kaiyuan Road, Luoyang 471003, China (Z.W.); (L.M.)
| | - Huili Wang
- University of North Carolina Hospitals, 101 Manning Dr, Chapel Hill, Orange County, NC 27599, USA;
| | - Yajie Guo
- Department of Emergency, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Jianxue Yang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China;
| |
Collapse
|
8
|
Akther T, McFadden WM, Zhang H, Kirby KA, Sarafianos SG, Wang Z. Design and Synthesis of New GS-6207 Subtypes for Targeting HIV-1 Capsid Protein. Int J Mol Sci 2024; 25:3734. [PMID: 38612545 PMCID: PMC11012105 DOI: 10.3390/ijms25073734] [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: 02/06/2024] [Revised: 03/08/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
HIV-1 capsid protein (CA) is the molecular target of the recently FDA-approved long acting injectable (LAI) drug lenacapavir (GS-6207). The quick emergence of CA mutations resistant to GS-6207 necessitates the design and synthesis of novel sub-chemotypes. We have conducted the structure-based design of two new sub-chemotypes combining the scaffold of GS-6207 and the N-terminal cap of PF74 analogs, the other important CA-targeting chemotype. The design was validated via induced-fit molecular docking. More importantly, we have worked out a general synthetic route to allow the modular synthesis of novel GS-6207 subtypes. Significantly, the desired stereochemistry of the skeleton C2 was confirmed via an X-ray crystal structure of the key synthetic intermediate 22a. Although the newly synthesized analogs did not show significant potency, our efforts herein will facilitate the future design and synthesis of novel subtypes with improved potency.
Collapse
Affiliation(s)
- Thamina Akther
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA;
| | - William M. McFadden
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (W.M.M.); (H.Z.)
| | - Huanchun Zhang
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (W.M.M.); (H.Z.)
| | - Karen A. Kirby
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (W.M.M.); (H.Z.)
| | - Stefan G. Sarafianos
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (W.M.M.); (H.Z.)
| | - Zhengqiang Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
9
|
Reddyrajula R, Etikyala U, Manga V, Kumar Dalimba U. Discovery of 1,2,3-triazole incorporated indole-piperazines as potent antitubercular agents: Design, synthesis, in vitro biological evaluation, molecular docking and ADME studies. Bioorg Med Chem 2024; 98:117562. [PMID: 38184947 DOI: 10.1016/j.bmc.2023.117562] [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: 10/19/2023] [Revised: 12/07/2023] [Accepted: 12/17/2023] [Indexed: 01/09/2024]
Abstract
In this report, a library consisting of three sets of indole-piperazine derivatives was designed through the molecular hybridization approach. In total, fifty new hybrid compounds (T1-T50) were synthesized and screened for antitubercular activity against Mycobacterium tuberculosis H37Rv strain (ATCC-27294). Five (T36, T43, T44, T48 and T49) among fifty compounds exhibited significant inhibitory potency with the MIC of 1.6 µg/mL, which is twofold more potent than the standard first-line TB drug Pyrazinamide and equipotent with Isoniazid. N-1,2,3-triazolyl indole-piperazine derivatives displayed improved inhibition activity as compared to the simple and N-benzyl indole-piperazine derivatives. In addition, the observed activity profile of indole-piperazines was similar to standard anti-TB drugs (isoniazid and pyrazinamide) against Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa strains, demonstrating the compounds' selectivity towards the Mycobacterium tuberculosis H37Rv strain. All the active anti-TB compounds are proved to be non-toxic (with IC50 > 300 μg/mL) as verified through the toxicity evaluation against VERO cell lines. Additionally, molecular docking studies against two target enzymes (Inh A and CYP121) were performed to validate the activity profile of indole-piperazine derivatives. Further, in silico-ADME prediction and pharmacokinetic parameters indicated that these compounds have good oral bioavailability.
Collapse
Affiliation(s)
- Rajkumar Reddyrajula
- Central Research facility, National Institute of Technology Karnataka, Surathkal, Mangalore 575025, India
| | - Umadevi Etikyala
- Medicinal Chemistry Laboratory, Department of Chemistry, Osmania University, Hyderabad 500076, India
| | - Vijjulatha Manga
- Medicinal Chemistry Laboratory, Department of Chemistry, Osmania University, Hyderabad 500076, India
| | - Udaya Kumar Dalimba
- Organic and Materials Chemistry Laboratory, Department of Chemistry, National Institute of Technology Karnataka, Surathkal, Mangalore 575025, India.
| |
Collapse
|
10
|
Farghaly TA, Masaret GS, Riyadh SM, Harras MF. A Literature Review Focusing on the Antiviral Activity of [1,2,4] and [1,2,3]-triazoles. Mini Rev Med Chem 2024; 24:1602-1629. [PMID: 38008942 DOI: 10.2174/0113895575277122231108095511] [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: 08/12/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 11/28/2023]
Abstract
Out of a variety of heterocycles, triazole scaffolds have been shown to play a significant part in a wide array of biological functions. Many drug compounds containing a triazole moiety with important antimicrobial, anticancer and antidepressant properties have been commercialized. In addition, the triazole scaffold exhibits remarkable antiviral activity either incorporated into nucleoside analogs or non-nucleosides. Many synthetic techniques have been produced by scientists around the world as a result of their wide-ranging biological function. In this review, we have tried to summarize new synthetic methods produced by diverse research groups as well as provide a comprehensive description of the function of [1,2,4] and [1,2,3]-triazole derivatives as antiviral agents. Antiviral triazole compounds have been shown to target a wide variety of molecular proteins. In addition, several strains of viruses, including the human immunodeficiency virus, SARS virus, hepatitis B and C viruses, influenza virus, Hantavirus, and herpes virus, were discovered to be susceptible to triazole derivatives. This review article covered the reports for antiviral activity of both 1,2,3- and 1,2,4-triazole moieties up to 2022.
Collapse
Affiliation(s)
- Thoraya A Farghaly
- Department of Chemistry, Faculty of Applied Science, Umm Al-Qura University, Makkah Almukaramah, 21514, Saudi Arabia
| | - Ghada S Masaret
- Department of Chemistry, Faculty of Applied Science, Umm Al-Qura University, Makkah Almukaramah, 21514, Saudi Arabia
| | - Sayed M Riyadh
- Chemistry Department, Faculty of Science, University of Cairo, Giza 12613, Egypt
| | - Marwa F Harras
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
11
|
ALSaeedy M, Al-Adhreai A, Alrabie A, Al-Qadsy I, Khamees HA, Alaizeri ZAM, Alhadlaq HA, Hasan A, Farooqui M. Novel Hybrid Triazoline - Triazole Glycosides: Synthesis, Characterization, Antimicrobial Activity study via In Vitro, and In Silico Means. Carbohydr Res 2023; 532:108877. [PMID: 37473676 DOI: 10.1016/j.carres.2023.108877] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/15/2023] [Accepted: 06/18/2023] [Indexed: 07/22/2023]
Abstract
Series of novel 1,2,3-triazole, and 1,2,3- triazoline glycosides (a-e) were efficiently synthesized starting from d-arabinose in an effort to synthesize a new type of hybrid molecules containing sugar azide. The key step involved is the introduction of a new group, ethylene glycol, to the anomeric site and protection of the hydroxyl groups with acetic anhydride. Following that, the acetyl group is converted into ethylene glycol to tosylate. Compound Azido ethyl-O-β-d-arabinofuranoside 4 was synthesized with good yield by treating the derivative 3 with sodium azide, which displaced the tosylate 3 and replaced it with the azide group. The new glycosides were synthesized via a 1,3-dipolar cycloaddition reaction between the intermediate compound 4 and several alkenes and alkynes. The triazole and triazoline compounds were characterized by FT-IR, 1H NMR, 13C NMR, LC/MS-IT-TOF spectral, and C·H.N. analysis. The antimicrobial screening was assayed using the disc diffusion technique revealed moderate to high potential inhibitory values against three test microorganisms compared to standard drugs. Their pharmacokinetics evaluation also showed promising drug-likeness and ADME properties. Furthermore, density functional theory (DFT) was utilized to obtain the molecular geometry of the title compounds utilizing B3LYP/6-311G++ (d, p), molecular electrostatic potential (MEP), frontier molecular orbitals (FMOs) through the investigation of HOMO and LUMO orbitals, and energy gap value. A lower energy gap value denotes that electrons can be transported more easily, indicating that molecule (b) is more reactive than other compounds. Molecular docking analysis revealed that all the designed triazole and triazoline glycosides interacted strongly inside the active site of the enzyme (PDB ID: 2Q85). and exhibits high docking scores, higher than the standard drug. The range of docking scores is -7.99 kcal/mol compound (a) to -7.42 kcal/mol compound (e).
Collapse
Affiliation(s)
- Mohammed ALSaeedy
- Department of Chemistry, Maulana Azad of Arts, Science and Commerce, Aurangabad, 431004, India; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey.
| | - Arwa Al-Adhreai
- Department of Chemistry, Maulana Azad of Arts, Science and Commerce, Aurangabad, 431004, India.
| | - Ali Alrabie
- Department of Chemistry, Maulana Azad of Arts, Science and Commerce, Aurangabad, 431004, India
| | - Inas Al-Qadsy
- Department of Chemistry, Maulana Azad of Arts, Science and Commerce, Aurangabad, 431004, India
| | - Hussien Ahmed Khamees
- Department of Studies in Physics, Manasagangotri, University of Mysore, Mysuru, 570006, Karnataka, India
| | - Zabn Allah M Alaizeri
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh-11451, Saudi Arabia
| | - Hisham A Alhadlaq
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh-11451, Saudi Arabia
| | - Ahmed Hasan
- Department of Pharmacology, Faculty of Pharmacy, University of Messina, 1-98122, Messina, Italy
| | - Mazahar Farooqui
- Department of Chemistry, Maulana Azad of Arts, Science and Commerce, Aurangabad, 431004, India
| |
Collapse
|
12
|
Zhang DW, Xu XS, Zhou R, Fu Z. Modulation of HIV-1 capsid multimerization by sennoside A and sennoside B via interaction with the NTD/CTD interface in capsid hexamer. Front Microbiol 2023; 14:1270258. [PMID: 37817748 PMCID: PMC10561090 DOI: 10.3389/fmicb.2023.1270258] [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: 07/31/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Small molecules that bind to the pocket targeted by a peptide, termed capsid assembly inhibitor (CAI), have shown antiviral effects with unique mechanisms of action. We report the discovery of two natural compounds, sennoside A (SA) and sennoside B (SB), derived from medicinal plants that bind to this pocket in the C-terminal domain of capsid (CA CTD). Both SA and SB were identified via a drug-screening campaign that utilized a time-resolved fluorescence resonance energy transfer assay. They inhibited the HIV-1 CA CTD/CAI interaction at sub-micromolar concentrations of 0.18 μM and 0.08 μM, respectively. Mutation of key residues (including Tyr 169, Leu 211, Asn 183, and Glu 187) in the CA CTD decreased their binding affinity to the CA monomer, from 1.35-fold to 4.17-fold. Furthermore, both compounds induced CA assembly in vitro and bound directly to the CA hexamer, suggesting that they interact with CA beyond the CA CTD. Molecular docking showed that both compounds were bound to the N-terminal domain (NTD)/CTD interface between adjacent protomers within the CA hexamer. SA established a hydrogen-bonding network with residues N57, V59, Q63, K70, and N74 of CA1-NTD and Q179 of CA2-CTD. SB formed hydrogen bonds with the N53, N70, and N74 residues of CA1-NTD, and the A177and Q179 residues of CA2-CTD. Both compounds, acting as glue, can bring αH4 in the NTD and αH9 in the CTD of the NTD/CTD interface close to each other. Collectively, our research indicates that SA and SB, which enhance CA assembly, could serve as novel chemical tools to identify agents that modulate HIV-1 CA assembly. These natural compounds may potentially lead to the development of new antiviral therapies with unique mechanisms of action.
Collapse
Affiliation(s)
- Da-Wei Zhang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Xiao-Shuang Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Rui Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiguo Fu
- Department of Orthopedics, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China
| |
Collapse
|
13
|
Majee S, Sarav M, Banik BK, Ray D. Recent Advances in the Green Synthesis of Active N-Heterocycles and Their Biological Activities. Pharmaceuticals (Basel) 2023; 16:873. [PMID: 37375820 DOI: 10.3390/ph16060873] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
N-heterocyclic scaffolds represent a privileged architecture in the process of drug design and development. It has widespread occurrence in synthetic and natural products, either those that are established or progressing as potent drug candidates. Additionally, numerous novel N-heterocyclic analogues with remarkable physiological significance and extended pharmaceutical applications are escalating progressively. Hence, the classical synthetic protocols need to be improvised according to modern requirements for efficient and eco-friendly approaches. Numerous methodologies and technologies emerged to address the green and sustainable production of various pharmaceutically and medicinally important N-heterocyclic compounds in last few years. In this context, the current review unveils greener alternatives for direct access to categorically differentiated N-heterocyclic derivatives and its application in the establishment of biologically active potent molecules for drug design. The green and sustainable methods accentuated in this review includes microwave-assisted reactions, solvent-free approaches, heterogeneous catalysis, ultrasound reactions, and biocatalysis.
Collapse
Affiliation(s)
- Suman Majee
- Amity Institute of Click Chemistry Research and Studies, Amity University, Sector 125, Noida 201313, Uttar Pradesh, India
- Amity Institute of Biotechnology, Amity University, Sector 125, Noida 201313, Uttar Pradesh, India
| | - Mansi Sarav
- Amity Institute of Biotechnology, Amity University, Sector 125, Noida 201313, Uttar Pradesh, India
| | - Bimal Krishna Banik
- Department of Mathematics and Natural Sciences, College of Sciences and Human Studies, Prince Mohammad Bin Fahd University, Al Khobar 31952, Saudi Arabia
| | - Devalina Ray
- Amity Institute of Click Chemistry Research and Studies, Amity University, Sector 125, Noida 201313, Uttar Pradesh, India
- Amity Institute of Biotechnology, Amity University, Sector 125, Noida 201313, Uttar Pradesh, India
| |
Collapse
|
14
|
Lahmadi G, Horchani M, Dbeibia A, Mahdhi A, Romdhane A, Lawson AM, Daïch A, Harrath AH, Ben Jannet H, Othman M. Novel Oleanolic Acid-Phtalimidines Tethered 1,2,3 Triazole Hybrids as Promising Antibacterial Agents: Design, Synthesis, In Vitro Experiments and In Silico Docking Studies. Molecules 2023; 28:4655. [PMID: 37375209 DOI: 10.3390/molecules28124655] [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: 05/18/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
As part of the valorization of agricultural waste into bioactive compounds, a series of structurally novel oleanolic acid ((3β-hydroxyolean-12-en-28-oic acid, OA-1)-phtalimidines (isoindolinones) conjugates 18a-u bearing 1,2,3-triazole moieties were designed and synthesized by treating an azide 4 previously prepared from OA-1 isolated from olive pomace (Olea europaea L.) with a wide range of propargylated phtalimidines using the Cu(I)-catalyzed click chemistry approach. OA-1 and its newly prepared analogues, 18a-u, were screened in vitro for their antibacterial activity against two Gram-positive bacteria, Staphylococcus aureus and Listeria monocytogenes, and two Gram-negative bacteria, Salmonella thyphimurium and Pseudomonas aeruginosa. Attractive results were obtained, notably against L. monocytogenes. Compounds 18d, 18g, and 18h exhibited the highest antibacterial activity when compared with OA-1 and other compounds in the series against tested pathogenic bacterial strains. A molecular docking study was performed to explore the binding mode of the most active derivatives into the active site of the ABC substrate-binding protein Lmo0181 from L. monocytogenes. Results showed the importance of both hydrogen bonding and hydrophobic interactions with the target protein and are in favor of the experimental data.
Collapse
Affiliation(s)
- Ghofrane Lahmadi
- Normandie University, URCOM, UNILEHAVRE, FR3021, UR 3221, 25 Rue Philippe Lebon, BP 540, F-76058 Le Havre, France
- Laboratory of Heterocyclic Chemistry, LR11ES39, Faculty of Science of Monastir, University of Monastir, Avenue of Environment, Monastir 5019, Tunisia
| | - Mabrouk Horchani
- Laboratory of Heterocyclic Chemistry, LR11ES39, Faculty of Science of Monastir, University of Monastir, Avenue of Environment, Monastir 5019, Tunisia
| | - Amal Dbeibia
- Laboratory of Analysis, Treatment and Valorization of Pollutants of the Environment and Products, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia
| | - Abdelkarim Mahdhi
- Laboratory of Analysis, Treatment and Valorization of Pollutants of the Environment and Products, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia
| | - Anis Romdhane
- Laboratory of Heterocyclic Chemistry, LR11ES39, Faculty of Science of Monastir, University of Monastir, Avenue of Environment, Monastir 5019, Tunisia
| | - Ata Martin Lawson
- Normandie University, URCOM, UNILEHAVRE, FR3021, UR 3221, 25 Rue Philippe Lebon, BP 540, F-76058 Le Havre, France
| | - Adam Daïch
- Normandie University, URCOM, UNILEHAVRE, FR3021, UR 3221, 25 Rue Philippe Lebon, BP 540, F-76058 Le Havre, France
| | - Abdel Halim Harrath
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hichem Ben Jannet
- Laboratory of Heterocyclic Chemistry, LR11ES39, Faculty of Science of Monastir, University of Monastir, Avenue of Environment, Monastir 5019, Tunisia
| | - Mohamed Othman
- Normandie University, URCOM, UNILEHAVRE, FR3021, UR 3221, 25 Rue Philippe Lebon, BP 540, F-76058 Le Havre, France
| |
Collapse
|
15
|
Moghimi P, Sabet-Sarvestani H, Shiri A. Synthesis, molecular docking and dynamics studies of pyridazino[4,5- b]quinoxalin-1(2 H)-ones as targeting main protease of COVID-19. J Biomol Struct Dyn 2023; 41:13198-13210. [PMID: 36951505 DOI: 10.1080/07391102.2023.2191127] [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: 06/21/2022] [Accepted: 01/15/2023] [Indexed: 03/24/2023]
Abstract
The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has created a crisis in public health. Because, the 3CLpro, the main protease of SARS-CoV-2, possesses a critical role in coronavirus replication, many efforts have been devoted to developing various inhibitors to prevent the fast spread of COVID-19. In the current work, a number of various pyridazino[4,5-b]quinoxalin-1(2H)-one derivatives bearing thiadiazine and thiadiazole fragments has been prepared via a straightforward and practical strategy involving the reaction of 2-(ethoxycarbonyl)-3-formylquinoxaline 1,4-dioxide with thiocarbohydrazide under reflux conditions. To determine the bioavailability of pyridazino[4,5-b]quinoxalin-1(2H)-one derivatives, Lipinski's rule of five has been carried out. Regarding this rule, none of the synthesized compounds exhibit any deviation from Lipinski's rule of five. Furthermore, molecular docking and molecular dynamics approaches have been implemented to figure out the potential interactions of products with SARS-CoV-2 main protease. The outcomes of molecular docking studies demonstrate that the phenyl and nitrophenyl substituted pyridazino[4,5-b]quinoxalin-1(2H)-one show the lowest binding affinity among the other compounds, indicating a favorable orientation in the active site of the chymotrypsin-like cysteine protease. In addition, the MD simulation performed to evaluate the stability of the protein-ligand complex represents that the average binding energy of the nitrophenyl complex is less than that of the phenyl complex. Therefore, according to the in silico results, the inhibitory effect of the nitrophenyl complex is more significant than the phenyl complex.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Parvin Moghimi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - Ali Shiri
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
16
|
Shah BM, Modi P, Trivedi P. Recent Investigation on Synthetic ‘Triazoles’ Scaffold as Potential Pharmacological Agents: A Comprehensive Survey. CHEMISTRY AFRICA 2023. [DOI: 10.1007/s42250-023-00617-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
17
|
Zhang DW, Xie L, Xu XS, Li Y, Xu X. A Broad-Spectrum Antiviral Molecule, Protoporphyrin IX, Acts as a Moderator of HIV-1 Capsid Assembly by Targeting the Capsid Hexamer. Microbiol Spectr 2023; 11:e0266322. [PMID: 36475726 PMCID: PMC9927277 DOI: 10.1128/spectrum.02663-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The capsid protein (CA), an essential component of human immunodeficiency virus type 1 (HIV-1), represents an appealing target for antivirals. Small molecules targeting the CAI-binding cavity in the C-terminal domain of HIV-1 CA (CA CTD) confer potent antiviral activities. In this study, we report that a small molecule, protoporphyrin IX (PPIX), targets the HIV-1 CA by binding to this pocket. PPIX was identified via in vitro drug screening, using a homogeneous and time-resolved fluorescence-based assay. CA multimerization and a biolayer interferometry (BLI) assay showed that PPIX promoted CA multimerization and bound directly to CA. The binding model of PPIX to CA CTD revealed that PPIX forms hydrogen bonds with the L211and E212 residues in the CA CTD. Moreover, the BLI assay demonstrated that this compound preferentially binds to the CA hexamer versus the monomer. The superposition of the CAI CTD-PPIX complex and the hexameric CA structure suggests that PPIX binds to the interface formed by the NTD and the CTD between adjacent protomers in the CA hexamer via the T72 and E212 residues, serving as a glue to enhance the multimerization of CA. Taken together, our studies demonstrate that PPIX, a hexamer-targeted CA assembly enhancer, should be a new chemical probe for the discovery of modulators of the HIV-1 capsid assembly. IMPORTANCE CA and its assembled viral core play essential roles in distinct steps during HIV-1 replication, including reverse transcription, integration, nuclear entry, virus assembly, and maturation through CA-CA or CA-host factor interactions. These functions of CA are fundamental for HIV-1 pathogenesis, making it an appealing target for antiviral therapy. In the present study, we identified protoporphyrin IX (PPIX) as a candidate CA modulator that can promote CA assembly and prefers binding the CA hexamer versus the monomer. PPIX, like a glue, bound at the interfaces between CA subunits to accelerate CA multimerization. Therefore, PPIX could be used as a new lead for a CA modulator, and it holds potential research applications.
Collapse
Affiliation(s)
- Da-Wei Zhang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Liangxu Xie
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Xiao-Shuang Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Yimin Li
- College of Pharmacy and Key Laboratory for Research and Development of “Qin Medicine” of Shaanxi Administration of Chinese Medicine, Shaanxi University of Chinese Medicine, Xixian New District, China
| | - Xiaojun Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| |
Collapse
|
18
|
Ding D, Xu S, Zhang X, Jiang X, Cocklin S, Dick A, Zhan P, Liu X. The discovery and design of novel HIV-1 capsid modulators and future perspectives. Expert Opin Drug Discov 2023; 18:5-12. [PMID: 36480372 DOI: 10.1080/17460441.2023.2157401] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Although combination antiretroviral therapy (cART) has achieved significant success in treating HIV, the emergence of multidrug-resistant viruses and cumulative medication toxicity make it necessary to find new classes of antiretroviral agents with novel mechanisms of action. With high sequence conservation, the HIV-1 capsid (CA) protein has attracted attention as a prospective therapeutic target due to its crucial structural and regulatory functions in the HIV-1 replication cycle. AREA COVERED Herein, the authors provide a cutting-edge overview of current advances in the design and discovery of CA modulators, PF74, GS-6207 and their derivativeswhich targets a therapeutically attractive NTD-CTD interprotomer pocket within the hexameric configuration of HIV-1 CA. The discovery and development of these compounds, and derivatives thereof, have provided valuable information for the design of second-generation CA-targeting antivirals. EXPERT OPINION Despite some successes in designing and discovering HIV-1 CA modulators, more studies are required to decipher which chemical groups confer specific desirable properties. The future of CA-modulating compounds may lie in covalent inhibition and the creation of proteolysis-targeting chimeras (PROTACs). Moreover, biological interrogation of the process of CA uncoating, virus-host interactions, and studies on the lattice-binding restriction factors may improve our knowledge of HIV-1 CA and support the design of new antiviral agents.
Collapse
Affiliation(s)
- Dang Ding
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Shujing Xu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xujie Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xiangyi Jiang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Simon Cocklin
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Alexej Dick
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
19
|
Xu S, Sun L, Zalloum WA, Huang T, Zhang X, Ding D, Shao X, Jiang X, Zhao F, Cocklin S, De Clercq E, Pannecouque C, Dick A, Liu X, Zhan P. Discovery and Mechanistic Investigation of Piperazinone Phenylalanine Derivatives with Terminal Indole or Benzene Ring as Novel HIV-1 Capsid Modulators. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238415. [PMID: 36500508 PMCID: PMC9739877 DOI: 10.3390/molecules27238415] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022]
Abstract
HIV-1 capsid (CA) performs multiple roles in the viral life cycle and is a promising target for antiviral development. In this work, we describe the design, synthesis, assessment of antiviral activity, and mechanistic investigation of 20 piperazinone phenylalanine derivatives with a terminal indole or benzene ring. Among them, F2-7f exhibited moderate anti-HIV-1 activity with an EC50 value of 5.89 μM, which was slightly weaker than the lead compound PF74 (EC50 = 0.75 μM). Interestingly, several compounds showed a preference for HIV-2 inhibitory activity, represented by 7f with an HIV-2 EC50 value of 4.52 μM and nearly 5-fold increased potency over anti-HIV-1 (EC50 = 21.81 μM), equivalent to PF74 (EC50 = 4.16 μM). Furthermore, F2-7f preferred to bind to the CA hexamer rather than to the monomer, similar to PF74, according to surface plasmon resonance results. Molecular dynamics simulation indicated that F2-7f and PF74 bound at the same site. Additionally, we computationally analyzed the ADMET properties for 7f and F2-7f. Based on this analysis, 7f and F2-7f were predicted to have improved drug-like properties and metabolic stability over PF74, and no toxicities were predicted based on the chemotype of 7f and F2-7f. Finally, the experimental metabolic stability results of F2-7f in human liver microsomes and human plasma moderately correlated with our computational prediction. Our findings show that F2-7f is a promising small molecule targeting the HIV-1 CA protein with considerable development potential.
Collapse
Affiliation(s)
- Shujing Xu
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Lin Sun
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Waleed A. Zalloum
- Department of Pharmacy, Faculty of Health Science, American University of Madaba, P.O. Box 2882, Amman 11821, Jordan
| | - Tianguang Huang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Xujie Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Dang Ding
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Xiaoyu Shao
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Xiangyi Jiang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Fabao Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Simon Cocklin
- Specifica Inc., The Santa Fe Railyard, 1607 Alcaldesa Street, Santa Fe, NM 87501, USA
| | - Erik De Clercq
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, K.U. Leuven, Herestraat 49 Postbus 1043 (09.A097), B-3000 Leuven, Belgium
| | - Christophe Pannecouque
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, K.U. Leuven, Herestraat 49 Postbus 1043 (09.A097), B-3000 Leuven, Belgium
- Correspondence: (C.P.); (A.D.); (X.L.); (P.Z.)
| | - Alexej Dick
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
- Correspondence: (C.P.); (A.D.); (X.L.); (P.Z.)
| | - Xinyong Liu
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
- Correspondence: (C.P.); (A.D.); (X.L.); (P.Z.)
| | - Peng Zhan
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, Jinan 250012, China
- Correspondence: (C.P.); (A.D.); (X.L.); (P.Z.)
| |
Collapse
|
20
|
Synthesis of Novel Ferrocene-Benzofuran Hybrids via Palladium- and Copper-Catalyzed Reactions. INORGANICS 2022. [DOI: 10.3390/inorganics10110205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The combination of the ferrocene skeleton with pharmacophores often leads to molecules with interesting biological properties. Five ferrocene-benzofuran hybrids of different structures were synthesized by transition metal catalyzed reactions. The efficiency of both homogeneous and heterogeneous catalytic methods was tested. The products were characterized using 1H, 13C NMR and FTIR spectroscopy, HRMS and cyclic voltammetry. The structure of one of the new compounds was also proved with X-ray crystallography. The new hybrids showed moderate cytotoxicity on MCF-7 and MDA-MB-231 cell lines. It is remarkable that the less curable MDA-MB-231 cell line was more sensitive to treatment with three ferrocene derivatives.
Collapse
|
21
|
Silver-Catalyzed Cascade Cyclization of Amino-NH-1,2,3-Triazoles with 2-Alkynylbenzaldehydes: An Access to Pentacyclic Fused Triazoles. Molecules 2022; 27:molecules27217567. [PMID: 36364393 PMCID: PMC9655256 DOI: 10.3390/molecules27217567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
An operationally simple Ag(I)-catalyzed approach for the synthesis of isoquinoline and quinazoline fused 1,2,3-triazoles was developed by a condensation and amination cyclization cascade of amino-NH-1,2,3-triazoles with 2-alkynylbenzaldehydes involving three new C-N bond formations in one manipulation, in which the group of -NH of the triazole ring serves as a nucleophile to form the quinazoline skeleton. The efficient protocol can be applied to a variety of substrates containing a range of functional groups, delivering novel pentacyclic fused 1,2,3-triazoles in good-to-excellent yields.
Collapse
|
22
|
Lengerli D, Ibis K, Nural Y, Banoglu E. The 1,2,3-triazole 'all-in-one' ring system in drug discovery: a good bioisostere, a good pharmacophore, a good linker, and a versatile synthetic tool. Expert Opin Drug Discov 2022; 17:1209-1236. [PMID: 36164263 DOI: 10.1080/17460441.2022.2129613] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The 1,2,3-triazole ring occupies an important space in medicinal chemistry due to its unique structural properties, synthetic versatility and pharmacological potential making it a critical scaffold. Since it is readily available through click chemistry for creating compound collections against various diseases, it has become an emerging area of interest for medicinal chemists. AREAS COVERED This review article addresses the unique properties of the1,2,3-triazole nucleus as an intriguing ring system in drug discovery while focusing on the most recent medicinal chemistry strategies exploited for the design and development of 1,2,3-triazole analogs as inhibitors of various biological targets. EXPERT OPINION Evidently, the 1,2,3-triazole ring with unique structural features has enormous potential in drug design against various diseases as a pharmacophore, a bioisoster or a structural platform. The most recent evidence indicates that it may be more emerging in drug molecules in near future along with an increasing understanding of its prominent roles in drug structures. The synthetic feasibility and versatility of triazole chemistry make it certainly ideal for creating compound libraries for more constructive structure-activity relationship studies. However, more comparative and target-specific studies are needed to gain a deeper understanding of the roles of the 1,2,3-triazole ring in molecular recognition.[Figure: see text].
Collapse
Affiliation(s)
- Deniz Lengerli
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Kübra Ibis
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Yahya Nural
- Department of Analytical Chemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Erden Banoglu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| |
Collapse
|
23
|
Yadav P, Kaushik C, Kumar M, Kumar A. Phthalimide/Naphthalimide containing 1,2,3-triazole hybrids: Synthesis and Antimicrobial Evaluation. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
24
|
Ji X, Li J, Sharma PP, Jiang X, Rathi B, Gao Z, Hu L, Kang D, De Clercq E, Cocklin S, Liu C, Pannecouque C, Dick A, Liu X, Zhan P. Design, Synthesis and Structure-Activity Relationships of Phenylalanine-Containing Peptidomimetics as Novel HIV-1 Capsid Binders Based on Ugi Four-Component Reaction. Molecules 2022; 27:molecules27185995. [PMID: 36144727 PMCID: PMC9502897 DOI: 10.3390/molecules27185995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/09/2022] [Accepted: 09/10/2022] [Indexed: 11/16/2022] Open
Abstract
As a key structural protein, HIV capsid (CA) protein plays multiple roles in the HIV life cycle, and is considered a promising target for anti-HIV treatment. Based on the structural information of CA modulator PF-74 bound to HIV-1 CA hexamer, 18 novel phenylalanine derivatives were synthesized via the Ugi four-component reaction. In vitro anti-HIV activity assays showed that most compounds exhibited low-micromolar-inhibitory potency against HIV. Among them, compound I-19 exhibited the best anti-HIV-1 activity (EC50 = 2.53 ± 0.84 μM, CC50 = 107.61 ± 27.43 μM). In addition, I-14 displayed excellent HIV-2 inhibitory activity (EC50 = 2.30 ± 0.11 μM, CC50 > 189.32 μM) with relatively low cytotoxicity, being more potent than that of the approved drug nevirapine (EC50 > 15.02 μM, CC50 > 15.2 μM). Additionally, surface plasmon resonance (SPR) binding assays demonstrated direct binding to the HIV CA protein. Moreover, molecular docking and molecular dynamics simulations provided additional information on the binding mode of I-19 to HIV-1 CA. In summary, we further explored the structure—activity relationships (SARs) and selectivity of anti-HIV-1/HIV-2 of PF-74 derivatives, which is conducive to discovering efficient anti-HIV drugs.
Collapse
Affiliation(s)
- Xiangkai Ji
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Jing Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Prem Prakash Sharma
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi 110007, India
| | - Xiangyi Jiang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Department of Chemistry, Hansraj College, University of Delhi, Delhi 110007, India
| | - Zhen Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Lide Hu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
- China-Belgium Collaborative Research Center for Innovative Antiviral Drugs of Shandong Province, 44 West Culture Road, Jinan 250012, China
| | - Erik De Clercq
- Laboratory of Virology and Chemotherapym, Rega Institute for Medical Research, K.U. Leuven, Herestraat 49 Postbus 1043 (09.A097), B-3000 Leuven, Belgium
| | - Simon Cocklin
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Chuanfeng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Christophe Pannecouque
- Laboratory of Virology and Chemotherapym, Rega Institute for Medical Research, K.U. Leuven, Herestraat 49 Postbus 1043 (09.A097), B-3000 Leuven, Belgium
- Correspondence: (C.P.); (A.D.); (X.L.); (P.Z.)
| | - Alexej Dick
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19104, USA
- Correspondence: (C.P.); (A.D.); (X.L.); (P.Z.)
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
- China-Belgium Collaborative Research Center for Innovative Antiviral Drugs of Shandong Province, 44 West Culture Road, Jinan 250012, China
- Correspondence: (C.P.); (A.D.); (X.L.); (P.Z.)
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
- China-Belgium Collaborative Research Center for Innovative Antiviral Drugs of Shandong Province, 44 West Culture Road, Jinan 250012, China
- Correspondence: (C.P.); (A.D.); (X.L.); (P.Z.)
| |
Collapse
|
25
|
Çot A, Çeşme M, Onur S, Aksakal E, Şahin İ, Tümer F. Rational design of 1,2,3-triazole hybrid structures as novel anticancer agents: synthesis, biological evaluation and molecular docking studies. J Biomol Struct Dyn 2022:1-9. [PMID: 35983627 DOI: 10.1080/07391102.2022.2112620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
New hybrid compounds belonging to the class of 1,4-disubstituted 1,2,3-triazoles were synthesized. The structural characterization of the synthesized compounds was performed using IR, 1H-NMR, 13C NMR and elemental analysis techniques. Diarylketones 1a and 1b were used as starting compounds for the synthesis of triazoles. The corresponding diarylmethanol derivatives (2a,b) were obtained from reduction of ketone units with NaBH4. Oxyalkynes (3a,b) were obtained by treating the hydroxyl group with NaH in anhydrous THF and then with propargylbromide. The target hybrid structures 6a-n were obtained from the metal-catalyzed "click reaction" of the arylazide and alkyne units. The newly synthesized compounds were structurally analysed using 1H-NMR, 13C-NMR, elemental analysis, LC-MS and FT-IR. The antioxidant and anticancer activities of all compounds were investigated. It has been determined that the new hybrid structures have very good antioxidant and anticancer activities according to the standards. In particular, compounds 6b, 6h, 6i and 6j (IC50: 1.87, 12.5, 7.22, 8.04 µM) showed excellent activity compared to standard 5-Fu (IC50: 40.89 µM). According to the results of molecular docking of compounds 6b and 6i with the highest cancer activity, MetAP-2 was found to have a high affinity through exposed polar and apolar contacts with fundemental residues in the binding pocket.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Aynur Çot
- Department of Chemistry, Faculty of Art and Sciences, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Mustafa Çeşme
- Department of Chemistry, Faculty of Art and Sciences, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Sultan Onur
- Department of Chemistry, Faculty of Art and Sciences, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Elif Aksakal
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ataturk University, Erzurum, Turkey
| | - İrfan Şahin
- Department of Chemistry, Faculty of Art and Sciences, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| | - Ferhan Tümer
- Department of Chemistry, Faculty of Art and Sciences, Kahramanmaras Sutcu Imam University, Kahramanmaras, Turkey
| |
Collapse
|
26
|
Yadav M, Lal K, Kumar A, Kumar A, Kumar D. Indole-chalcone linked 1,2,3-triazole hybrids: Facile synthesis, antimicrobial evaluation and docking studies as potential antimicrobial agents. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
27
|
Holanda VN, Lima EMDA, da Silva WV, Maia RT, Medeiros RDL, Ghosh A, Lima VLDM, de Figueiredo RCBQ. Identification of 1,2,3-triazole-phthalimide derivatives as potential drugs against COVID-19: a virtual screening, docking and molecular dynamic study. J Biomol Struct Dyn 2022; 40:5462-5480. [PMID: 33459182 PMCID: PMC7832388 DOI: 10.1080/07391102.2020.1871073] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/28/2020] [Indexed: 01/07/2023]
Abstract
In this work we aimed to perform an in silico predictive screening, docking and molecular dynamic study to identify 1,2,3-triazole-phthalimide derivatives as drug candidates against SARS-CoV-2. The in silico prediction of pharmacokinetic and toxicological properties of hundred one 1,2,3-triazole-phtalimide derivatives, obtained from SciFinder® library, were investigated. Compounds that did not show good gastrointestinal absorption, violated the Lipinski's rules, proved to be positive for the AMES test, and showed to be hepatotoxic or immunotoxic in our ADMET analysis, were filtered out of our study. The hit compounds were further subjected to molecular docking on SARS-CoV-2 target proteins. The ADMET analysis revealed that 43 derivatives violated the Lipinski's rules and 51 other compounds showed to be positive for the toxicity test. Seven 1,2,3-triazole-phthalimide derivatives (A7, A8, B05, E35, E38, E39, and E40) were selected for molecular docking and MFCC-ab initio analysis. The results of molecular docking pointed the derivative E40 as a promising compound interacting with multiple target proteins of SARS-CoV-2. The complex E40-Mpro was found to have minimum binding energy of -10.26 kcal/mol and a general energy balance, calculated by the quantum mechanical analysis, of -8.63 eV. MD simulation and MMGBSA calculations confirmed that the derivatives E38 and E40 have high binding energies of -63.47 ± 3 and -63.31 ± 7 kcal/mol against SARS-CoV-2 main protease. In addition, the derivative E40 exhibited excellent interaction values and inhibitory potential against SAR-Cov-2 main protease and viral nucleocapsid proteins, suggesting this derivative as a potent antiviral for the treatment and/or prophylaxis of COVID-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vanderlan Nogueira Holanda
- Laboratório de Lipídios e Aplicação de Biomoléculas em Doenças Prevalentes e Negligenciadas, Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
- Laboratório de Biologia Celular de Patógenos, Instituto Aggeu Magalhães, Departamento de Microbiologia, Instituto Aggeu Magalhães – IAM/FIOCRUZ-PE, Recife, Pernambuco, Brazil
| | - Elton Marlon de Araújo Lima
- Laboratório de Biologia Celular de Patógenos, Instituto Aggeu Magalhães, Departamento de Microbiologia, Instituto Aggeu Magalhães – IAM/FIOCRUZ-PE, Recife, Pernambuco, Brazil
- Laboratório de Polímeros Não-Convencionais, Departamento de Física, Centro de Ciências Exatas e da Natureza, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Welson Vicente da Silva
- Laboratório de Biologia Celular de Patógenos, Instituto Aggeu Magalhães, Departamento de Microbiologia, Instituto Aggeu Magalhães – IAM/FIOCRUZ-PE, Recife, Pernambuco, Brazil
| | - Rafael Trindade Maia
- Centro de Desenvolvimento Sustentável do Semiárido, Universidade Federal de Campina Grande, Sumé, Paraíba, Brazil
| | | | - Arabinda Ghosh
- Microbiology Division, Department of Botany, Gauhati University, Guwahati, Assam, India
| | - Vera Lúcia de Menezes Lima
- Laboratório de Lipídios e Aplicação de Biomoléculas em Doenças Prevalentes e Negligenciadas, Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Regina Celia Bressan Queiroz de Figueiredo
- Laboratório de Biologia Celular de Patógenos, Instituto Aggeu Magalhães, Departamento de Microbiologia, Instituto Aggeu Magalhães – IAM/FIOCRUZ-PE, Recife, Pernambuco, Brazil
| |
Collapse
|
28
|
Mirdarvatan V, Bahramian B, Khalaji AD, Bakherad M, Charles C, Gómez‐García CJ, Rezaeifard A, Triki S. Part‐per‐million catalysis of azide‐alkyne cycloaddition reaction in water using a new ferromagnetic μ
1,1
‐N
3
bridged dinuclear Cu (II) complex. Appl Organomet Chem 2022. [DOI: 10.1002/aoc.6841] [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)
- Vahid Mirdarvatan
- Department of Chemistry Shahrood University of Technology Shahrood Iran
| | - Bahram Bahramian
- Department of Chemistry Shahrood University of Technology Shahrood Iran
| | | | - Mohammad Bakherad
- Department of Chemistry Shahrood University of Technology Shahrood Iran
| | - Catherine Charles
- Univ Brest, CNRS, CEMCA, 6 Avenue Victor Le Gorgeu, C.S. 93837‐29238 Brest France
| | - Carlos J. Gómez‐García
- Departamento de Química Inorgánica. Universidad de Valencia, C/Dr. Moliner 50. 46100 Burjasot Spain
| | - Amin Rezaeifard
- Department of Chemistry Shahrood University of Technology Shahrood Iran
| | - Smail Triki
- Univ Brest, CNRS, CEMCA, 6 Avenue Victor Le Gorgeu, C.S. 93837‐29238 Brest France
| |
Collapse
|
29
|
De Masi R, Orlando S. GANAB and N-Glycans Substrates Are Relevant in Human Physiology, Polycystic Pathology and Multiple Sclerosis: A Review. Int J Mol Sci 2022; 23:7373. [PMID: 35806376 PMCID: PMC9266668 DOI: 10.3390/ijms23137373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Glycans are one of the four fundamental macromolecular components of living matter, and they are highly regulated in the cell. Their functions are metabolic, structural and modulatory. In particular, ER resident N-glycans participate with the Glc3Man9GlcNAc2 highly conserved sequence, in protein folding process, where the physiological balance between glycosylation/deglycosylation on the innermost glucose residue takes place, according GANAB/UGGT concentration ratio. However, under abnormal conditions, the cell adapts to the glucose availability by adopting an aerobic or anaerobic regimen of glycolysis, or to external stimuli through internal or external recognition patterns, so it responds to pathogenic noxa with unfolded protein response (UPR). UPR can affect Multiple Sclerosis (MS) and several neurological and metabolic diseases via the BiP stress sensor, resulting in ATF6, PERK and IRE1 activation. Furthermore, the abnormal GANAB expression has been observed in MS, systemic lupus erythematous, male germinal epithelium and predisposed highly replicating cells of the kidney tubules and bile ducts. The latter is the case of Polycystic Liver Disease (PCLD) and Polycystic Kidney Disease (PCKD), where genetically induced GANAB loss affects polycystin-1 (PC1) and polycystin-2 (PC2), resulting in altered protein quality control and cyst formation phenomenon. Our topics resume the role of glycans in cell physiology, highlighting the N-glycans one, as a substrate of GANAB, which is an emerging key molecule in MS and other human pathologies.
Collapse
Affiliation(s)
- Roberto De Masi
- Complex Operative Unit of Neurology, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy;
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| | - Stefania Orlando
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| |
Collapse
|
30
|
Ramesh D, Mohanty AK, De A, Vijayakumar BG, Sethumadhavan A, Muthuvel SK, Mani M, Kannan T. Uracil derivatives as HIV-1 capsid protein inhibitors: design, in silico, in vitro and cytotoxicity studies. RSC Adv 2022; 12:17466-17480. [PMID: 35765450 PMCID: PMC9190787 DOI: 10.1039/d2ra02450k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/29/2022] [Indexed: 11/24/2022] Open
Abstract
A series of novel uracil derivatives such as bispyrimidine dione and tetrapyrimidine dione derivatives were designed based on the existing four-point pharmacophore model as effective HIV capsid protein inhibitors. The compounds were initially docked with an HIV capsid protein monomer to rationalize the ideas of design and to find the potential binding modes. The successful design and computational studies led to the synthesis of bispyrimidine dione and tetrapyrimidine dione derivatives from uracil and aromatic aldehydes in the presence of HCl using novel methodology. The in vitro evaluation in HIV p24 assay revealed five potential uracil derivatives with IC50 values ranging from 191.5 μg ml−1 to 62.5 μg ml−1. The meta-chloro substituted uracil compound 9a showed promising activity with an IC50 value of 62.5 μg ml−1 which is well correlated with the computational studies. As expected, all the active compounds were noncytotoxic in BA/F3 and Mo7e cell lines highlighting the thoughtful design. The structure activity relationship indicates the position priority and lower log P values as the possible cause of inhibitory potential of the uracil compounds. The paper describes the design, synthesis, computational and biological validation of a series of novel uracil derivatives as effective HIV capsid protein inhibitors.![]()
Collapse
Affiliation(s)
- Deepthi Ramesh
- Department of Chemistry, Pondicherry University Kalapet Puducherry-605014 India
| | - Amaresh Kumar Mohanty
- Department of Bioinformatics, Pondicherry University Kalapet Puducherry-605014 India
| | - Anirban De
- Department of Chemistry, Pondicherry University Kalapet Puducherry-605014 India
| | | | | | - Suresh Kumar Muthuvel
- Department of Bioinformatics, Pondicherry University Kalapet Puducherry-605014 India
| | - Maheswaran Mani
- Department of Microbiology, Pondicherry University Kalapet Puducherry-605014 India
| | | |
Collapse
|
31
|
Verma N, Bera S, Gonnade R, Mondal D. Regioselective synthesis of 1,4,5‐Trisubstituted‐1,2,3‐Triazole Derivatives from α,β‐Unsaturated Carbonyls. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200317] [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)
- Naimish Verma
- Central University of Gujarat School of Chemical Sciences Sector-30 382030 Gandhinagar INDIA
| | - Smritilekha Bera
- Central University of Gujarat School of Chemical Sciences 30 sector 382030 Gandhinagar INDIA
| | - Rajesh Gonnade
- CSIR-National Chemical Laboratory: National Chemical Laboratory CSIR Physical and Materials Chemistry Division 411008 Pune INDIA
| | - Dhananjoy Mondal
- Central University of Gujarat School of Chemical Sciences Sector-30 382030 Gandhinagar INDIA
| |
Collapse
|
32
|
Cu2O/CuO@mont K 10 promoted one-pot synthesis of 1,2,3-triazoles through azide-alkyne cycloaddition reaction. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-022-02203-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
33
|
Popović-Djordjević J, Quispe C, Giordo R, Kostić A, Katanić Stanković JS, Tsouh Fokou PV, Carbone K, Martorell M, Kumar M, Pintus G, Sharifi-Rad J, Docea AO, Calina D. Natural products and synthetic analogues against HIV: A perspective to develop new potential anti-HIV drugs. Eur J Med Chem 2022; 233:114217. [DOI: 10.1016/j.ejmech.2022.114217] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/13/2022] [Accepted: 02/20/2022] [Indexed: 12/22/2022]
|
34
|
Pingaew R, Choomuenwai V, Leechaisit R, Prachayasittikul V, Prachayasittikul S, Prachayasittikul V. 1,2,3-Triazole Scaffold in Recent Medicinal Applications: Synthesis and Anticancer Potentials. HETEROCYCLES 2022. [DOI: 10.3987/rev-22-sr(r)4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
35
|
McFadden WM, Snyder AA, Kirby KA, Tedbury PR, Raj M, Wang Z, Sarafianos SG. Rotten to the core: antivirals targeting the HIV-1 capsid core. Retrovirology 2021; 18:41. [PMID: 34937567 PMCID: PMC8693499 DOI: 10.1186/s12977-021-00583-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/09/2021] [Indexed: 12/20/2022] Open
Abstract
The capsid core of HIV-1 is a large macromolecular assembly that surrounds the viral genome and is an essential component of the infectious virus. In addition to its multiple roles throughout the viral life cycle, the capsid interacts with multiple host factors. Owing to its indispensable nature, the HIV-1 capsid has been the target of numerous antiretrovirals, though most capsid-targeting molecules have not had clinical success until recently. Lenacapavir, a long-acting drug that targets the HIV-1 capsid, is currently undergoing phase 2/3 clinical trials, making it the most successful capsid inhibitor to-date. In this review, we detail the role of the HIV-1 capsid protein in the virus life cycle, categorize antiviral compounds based on their targeting of five sites within the HIV-1 capsid, and discuss their molecular interactions and mechanisms of action. The diverse range of inhibition mechanisms provides insight into possible new strategies for designing novel HIV-1 drugs and furthers our understanding of HIV-1 biology. ![]()
Collapse
Affiliation(s)
- William M McFadden
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Alexa A Snyder
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Karen A Kirby
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Philip R Tedbury
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Monika Raj
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Zhengqiang Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Stefan G Sarafianos
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA.
| |
Collapse
|
36
|
Sahani RL, Akther T, Cilento ME, Castaner AE, Zhang H, Kirby KA, Xie J, Sarafianos SG, Wang Z. Potency and metabolic stability: a molecular hybrid case in the design of novel PF74-like small molecules targeting HIV-1 capsid protein. RSC Med Chem 2021; 12:2031-2044. [PMID: 35028563 DOI: 10.1039/d1md00292a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/27/2021] [Indexed: 12/27/2022] Open
Abstract
PF74 (1) is a potent and well-characterized prototypical small molecule targeting human immunodeficiency virus type 1 (HIV-1) capsid protein (CA), but not a viable antiviral lead due to the lack of metabolic stability. We report herein our molecular hybridization-based medicinal chemistry efforts toward potent and metabolically stable PF74-like small molecules. The design of the new sub-chemotype 4 rationally combines binding features of two recently reported PF74-like compounds 2 and 3. The subsequent confirmation and structure-activity relationship (SAR) of hit 4a entailed the chemical synthesis of 37 novel analogs, most of which showed modest but meaningful thermal shift, and low μM antiviral activity. The most potent analogs (4a, 4d, 4o, and 4r) all exhibited noticeably improved metabolic stability over PF74. Molecular modeling suggests that these new analogs bind to the PF74 binding site. Overall, our work demonstrated that the molecular hybridization approach is suitable for designing compounds with balanced potency and metabolic stability.
Collapse
Affiliation(s)
- Rajkumar Lalji Sahani
- Center for Drug Design, College of Pharmacy, University of Minnesota Minneapolis MN 55455 USA
| | - Thamina Akther
- Center for Drug Design, College of Pharmacy, University of Minnesota Minneapolis MN 55455 USA
| | - Maria E Cilento
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine Atlanta GA 30322 USA
| | - Andres Emanuelli Castaner
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine Atlanta GA 30322 USA
| | - Huanchun Zhang
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine Atlanta GA 30322 USA
| | - Karen A Kirby
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine Atlanta GA 30322 USA.,Children's Healthcare of Atlanta Atlanta GA 30322 USA
| | - Jiashu Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota Minneapolis MN 55455 USA
| | - Stefan G Sarafianos
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine Atlanta GA 30322 USA.,Children's Healthcare of Atlanta Atlanta GA 30322 USA
| | - Zhengqiang Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota Minneapolis MN 55455 USA
| |
Collapse
|
37
|
Biological Evaluation of 4-(1H-triazol-1-yl)benzoic Acid Hybrids as Antioxidant Agents: In Vitro Screening and DFT Study. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112411642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Fourteen triazole benzoic acid hybrids were previously characterized. This work aimed to screen their in vitro antioxidant activity using different assays, i.e., DPPH (1,1-diphenyl-1-picrylhydrazyl), reducing the power capability, FRAP (ferric reducing antioxidants power) and ABTS (2,2′-azino-bis(3-ethylben zothiazoline-6-sulfonate) radical scavenging. The 14 compounds showed antioxidant properties in relation to standard BHA (butylated hydroxylanisole) and Trolox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid). Higher antioxidant activity was observed by the parent (1) at a concentration of 100 µg/mL (89.95 ± 0.34 and 88.59 ± 0.13%) when tested by DPPH and ABTS methods in relation to BHA at 100 µg/mL (95.02 ± 0.74 and 96.18 ± 0.33%). The parent (2) demonstrated remarkable scavenging activity when tested by ABTS (62.00 ± 0.24%), however, 3 was less active (29.98 ± 0.13%). Compounds 5, 6, 9, and 11 exhibited good scavenging activity compared to 1. DFT studies were performed using the B3LYP/6-311++g (2d,2p) level of theory to evaluate different antioxidant descriptors for the targets. Three antioxidant mechanisms, i.e., hydrogen atom transfer (HAT), sequential electron transfer proton transfer (SETPT) and sequential proton loss electron transfer (SPLET) were suggested to describe the antioxidant properties of 1–14. Out of the 14 triazole benzoic acid hybrids, 5, 9, 6, and 11 showed some good theoretical results, which were in agreement with some experimental outcomes. Based on the computed (PA and ETE) and (BDE and IP) values in (SPLET) and (HAT and SETPT) mechanisms, respectively, compound 9 emerged has having good antioxidant activity.
Collapse
|
38
|
Selim A, Neethu KM, Gowri V, Sartaliya S, Kaur S, Jayamurugan G. Thiol‐Functionalized Cellulose Wrapped Copperoxide as a Green Nano Catalyst for Regiospecific Azide‐Alkyne Cycloaddition Reaction: Application in Rufinamide Synthesis. ASIAN J ORG CHEM 2021. [DOI: 10.1002/ajoc.202100658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Abdul Selim
- Institute of Nano Science and Technology (INST) Knowledge City, Sector 81 Mohali Punjab 140306 India
| | - K. M. Neethu
- Institute of Nano Science and Technology (INST) Knowledge City, Sector 81 Mohali Punjab 140306 India
| | - Vijayendran Gowri
- Institute of Nano Science and Technology (INST) Knowledge City, Sector 81 Mohali Punjab 140306 India
| | - Shaifali Sartaliya
- Institute of Nano Science and Technology (INST) Knowledge City, Sector 81 Mohali Punjab 140306 India
| | - Sharanjeet Kaur
- Institute of Nano Science and Technology (INST) Knowledge City, Sector 81 Mohali Punjab 140306 India
| | - Govindasamy Jayamurugan
- Institute of Nano Science and Technology (INST) Knowledge City, Sector 81 Mohali Punjab 140306 India
| |
Collapse
|
39
|
Peters SJ, Patel K. Electron Distribution in 1,2,3-Benzotriazole and 1,2,3-Triazole Anion Radical Isomers: An EPR and DFT Study. J Org Chem 2021; 86:14786-14796. [PMID: 34633818 DOI: 10.1021/acs.joc.1c01584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The anion radicals of N1- and N2-alkylbenzotriazoles and alkyltriazoles (alkyl = methyl or isopropyl) have been generated by low-temperature potassium metal reduction in tetrahydrofuran. Electron paramagnetic resonance (EPR) analysis and density functional theory calculations reveal that the electron spin distribution within the triazole ring of these systems is markedly different. The magnitude of the electron-nitrogen couplings along with the calculated spin densities reveals that the N2-alkylbenzotriazole and N2-alkyltriazole anion radicals have significantly greater electron spin residing within the N3 portion of the triazole ring compared with that of the respective N1 isomers. These differences impact the overall geometry of the triazole ring where both N2-isomers lose planarity upon reduction. Experimental and computational results reveal that the N2-methyltriazole anion radical has the largest concentration of electron spin residing in the N3 moiety compared to that of the other three anion radicals studied. Significant anisotropic line broadening is observed in the EPR spectrum of the N2-methyltriazole anion radical, which is a consequence of the large nitrogen hyperfine couplings and sufficiently slow rotational motion of this species in solution.
Collapse
Affiliation(s)
- Steven J Peters
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| | - Krutil Patel
- Department of Chemistry, Illinois State University, Normal, Illinois 61790-4160, United States
| |
Collapse
|
40
|
El‐Sayed HA, Moustafa AH, Masry AA, Amer AM, Mohammed SM. An efficient synthesis of 4,6‐diarylnicotinonitrile‐acetamide hybrids via 1,2,3‐triazole linker as multitarget microbial inhibitors. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Hassan A. El‐Sayed
- Department of Chemistry, Faculty of Science Zagazig University Zagazig Egypt
| | - Ahmed H. Moustafa
- Department of Chemistry, Faculty of Science Zagazig University Zagazig Egypt
| | - Asmaa A. Masry
- Department of Chemistry, Faculty of Science Zagazig University Zagazig Egypt
| | - Atef M. Amer
- Department of Chemistry, Faculty of Science Zagazig University Zagazig Egypt
| | - Samar M. Mohammed
- Department of Chemistry, Faculty of Science Zagazig University Zagazig Egypt
| |
Collapse
|
41
|
Synthesis and biological evaluation of 1,6-bis-triazole-2,3,4-tri-O-benzyl-α-d-glucopyranosides as a novel α-glucosidase inhibitor in the treatment of Type 2 diabetes. Bioorg Med Chem Lett 2021; 50:128331. [PMID: 34418573 DOI: 10.1016/j.bmcl.2021.128331] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 11/21/2022]
Abstract
A novel series of 1,6-bis-triazole-benzyl-α-glucoside derivatives (7a-7ee) were designed, synthesized and evaluated for inhibitory activity against α-glucosidase. Most of the synthesized compounds exhibited good activity with IC50 ranging from 3.73 µM to 53.34 µM and are more potent than the standard drug acarbose (IC50 = 146.25 ± 0.40 µM). SARs study showed the ester and menthol moiety play an important role in the inhibitory activity. The molecular docking model of the potent compounds 7f, 7z, 7cc and 7dd showed good binding energy and interacts well with amino acid residues around the active site of the enzyme, which confirmed the in vitro activity results.
Collapse
|
42
|
Design, synthesis, and antiviral activity of phenylalanine derivatives as HIV-1 capsid inhibitors. Bioorg Med Chem 2021; 48:116414. [PMID: 34562701 DOI: 10.1016/j.bmc.2021.116414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 11/23/2022]
Abstract
The HIV-1 Capsid (CA) is considered as a promising target for the development of potent antiviral drugs, due to its multiple roles during the viral life cycle. Herein, we report the design, synthesis, and antiviral activity evaluation of series of novel phenylalanine derivatives as HIV-1 CA protein inhibitors. Among them, 4-methoxy-N-methylaniline substituted phenylalanine (II-13c) and indolin-5-amine substituted phenylalanine (V-25i) displayed exceptional anti-HIV-1 activity with the EC50 value of 5.14 and 2.57 μM respectively, which is slightly weaker than that of lead compound PF-74 (EC50 = 0.42 μM). Besides, surface plasmon resonance (SPR) binding assay demonstrated II-13c and V-25i prefer to combine with CA hexamer rather than monomer, which is similar to PF-74. Subsequently, molecular dynamics simulation (MD) revealed potential interactions between representative compounds with HIV-1 CA hexamer. Overall, this work laid a solid foundation for further structural optimization to discover novel promising HIV-1 CA inhibitors.
Collapse
|
43
|
Xu S, Sun L, Dick A, Zalloum WA, Huang T, Meuser ME, Zhang X, Tao Y, Cherukupalli S, Ding D, Ding X, Gao S, Jiang X, Kang D, De Clercq E, Pannecouque C, Cocklin S, Liu X, Zhan P. Design, synthesis, and mechanistic investigations of phenylalanine derivatives containing a benzothiazole moiety as HIV-1 capsid inhibitors with improved metabolic stability. Eur J Med Chem 2021; 227:113903. [PMID: 34653770 DOI: 10.1016/j.ejmech.2021.113903] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/03/2021] [Accepted: 10/03/2021] [Indexed: 12/17/2022]
Abstract
Further clinical development of PF74, a lead compound targeting HIV-1 capsid, is impeded by low antiviral activity and inferior metabolic stability. By modifying the benzene (region I) and indole of PF74, we identified two potent compounds (7m and 7u) with significantly improved metabolic stability. Compared to PF74, 7u displayed greater metabolic stability in human liver microsomes (HLMs) with half-life (t1/2) 109-fold that of PF74. Moreover, mechanism of action (MOA) studies demonstrated that 7m and 7u effectively mirrored the MOA of compounds that interact within the PF74 interprotomer pocket, showing direct and robust interactions with recombinant CA, and 7u displaying antiviral effects in both the early and late stages of HIV-1 replication. Furthermore, MD simulation corroborated that 7u was bound to the PF74 binding site, and the results of the online molinspiration software predicted that 7m and 7u had desirable physicochemical properties. Unexpectedly, this series of compounds exhibited better antiviral activity than PF74 against HIV-2, represented by compound 7m whose anti-HIV-2 activity was almost 5 times increased potency over PF74. Therefore, we have rationally redesigned the PF74 chemotype to inhibitors with novel structures and enhanced metabolic stability in this study. We hope that these new compounds can serve as a blueprint for developing a new generation of HIV treatment regimens.
Collapse
Affiliation(s)
- Shujing Xu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Lin Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Alexej Dick
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, PA, 19102, USA
| | - Waleed A Zalloum
- Department of Pharmacy, Faculty of Health Science, American University of Madaba, P.O Box 2882, Amman, 11821, Jordan
| | - Tianguang Huang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Megan E Meuser
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, PA, 19102, USA
| | - Xujie Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Yucen Tao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Srinivasulu Cherukupalli
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Dang Ding
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Xiao Ding
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Shenghua Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Xiangyi Jiang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China
| | - Erik De Clercq
- Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, K.U. Leuven, Herestraat 49 Postbus 1043 (09.A097), B-3000, Leuven, Belgium
| | - Christophe Pannecouque
- Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, K.U. Leuven, Herestraat 49 Postbus 1043 (09.A097), B-3000, Leuven, Belgium.
| | - Simon Cocklin
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, PA, 19102, USA.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China.
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012, Jinan, Shandong, PR China.
| |
Collapse
|
44
|
Vellingiri A, Murugan D, Gnana Kumar G, Alagusundaram P. An elegant and efficient synthesis of heterocycles integrated with
bis
‐
N
‐acyl
pyrazoline and
bis
‐1, 2,
3‐triazole
via a green synthetic methodology. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
| | - Dinesh Murugan
- School of Chemistry, Madurai Kamaraj University Madurai India
| | | | | |
Collapse
|
45
|
Abuelizz HA, Al-Salahi R. An overview of triazoloquinazolines: Pharmacological significance and recent developments. Bioorg Chem 2021; 115:105263. [PMID: 34426148 DOI: 10.1016/j.bioorg.2021.105263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/14/2021] [Accepted: 08/07/2021] [Indexed: 01/26/2023]
Abstract
Nitrogen heterocyclic rings have participated to constitute most of the drugs and several pharmacologically related compounds. The existence of such hetero atoms/groups in heterocyclic systems privileged specificities in their biological objectives. Particularly, quinazoline and triazole are biologically imperative platforms known to be linked with various pharmacological activities. Some of the prominent pharmacological responses ascribed to these systems are analgesic, antiinflammatory, anticonvulsant, hypnotic, antihistaminic, antihypertensive, anticancer, antimicrobial, antitubercular, antiviral and antimalarial activities. This diversity in the pharmacological outputs for both triazole and quinazoline systems has encouraged the medicinal chemistry researchers to create several chemical routes aiming at the incorporation of two rings in one molecule named triazoloquinazoline system. This system has shown multiple potential activities against numerous targets. Correlation the specific structural features of triazoloquinazoline system with its pharmacological purposes has successively been achieved by performing several pharmacological examinations and structure-activity relationship studies. The development of triazoloquinazoline derivatives and the understanding of their pharmacological targets offer opportunities for novel therapeutics. This review mainly emphases on the medicinal chemistry aspects of triazoloquinazolines including synthesis, reactivity, biological activity and structure activity relationship studies (SARs). Moreover, this review collates literature reported by researchers on triazoquinazolines and provides detailed attention on their analogs pharmacological activities in the perspective of drug development and discovery.
Collapse
Affiliation(s)
- Hatem A Abuelizz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, PO Box 2457, Riyadh 11451, Saudi Arabia
| | - Rashad Al-Salahi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, PO Box 2457, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
46
|
Talukdar R, Singh V, Mourya H, Nasibullah M, Tiwari B. Stitching Triazoles to Arenes via a Transition Metal-Free Aryne Diels-Alder/1,3-Prototropic Shift/Dehydrobromination Cascade. J Org Chem 2021; 86:12277-12284. [PMID: 34328329 DOI: 10.1021/acs.joc.1c00562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The 1,2,3-triazole-fused polyaromatic frameworks are traditionally obtained through transition metal-catalyzed C-H/C-X arylation of the preinstalled triazoles at a very high temperature. Herein, a transition metal-free direct synthesis via an aryne Diels-Alder/1,3-prototropic shift/dehydrobromination cascade is reported. This method gives access to triazole-fused aromatics as well as the corresponding dihydrocarbocycles under mild reaction conditions.
Collapse
Affiliation(s)
- Ranadeep Talukdar
- Division of Molecular Synthesis & Drug Discovery, Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow 226014, India
| | - Vikram Singh
- Division of Molecular Synthesis & Drug Discovery, Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow 226014, India
| | - Hemlata Mourya
- Division of Molecular Synthesis & Drug Discovery, Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow 226014, India
| | - Malik Nasibullah
- Department of Chemistry, Integral University, Dasauli, Bas-ha Kursi Road, Lucknow 226026, India
| | - Bhoopendra Tiwari
- Division of Molecular Synthesis & Drug Discovery, Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow 226014, India
| |
Collapse
|
47
|
Haroun M, Tratrat C, Kochkar H, Nair AB. CDATA[Recent Advances in the Development of 1,2,3-Triazole-containing Derivatives as Potential Antifungal Agents and Inhibitors of Lanoster ol 14α-Demethylase. Curr Top Med Chem 2021; 21:462-506. [PMID: 33319673 DOI: 10.2174/1568026621999201214232018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/12/2020] [Accepted: 11/03/2020] [Indexed: 11/22/2022]
Abstract
1,2,3-Triazole, a five-membered heterocyclic nucleus, is widely recognized as a key chromophore of great value in medicinal chemistry for delivering compounds possessing innumerable biological activities, including antimicrobial, antitubercular, antidiabetic, antiviral, antitumor, antioxidants, and anti-inflammatory activities. Mainly, in the past years, diverse conjugates carrying this biologically valuable core have been reported due to their attractive fungicidal potential and potent effects on various infective targets. Hence, hybridization of 1,2,3-triazole with other antimicrobial pharmacophores appears to be a judicious strategy to develop new effective anti-fungal candidates to combat the emergence of drug-sensitive and drug-resistant infectious diseases. Thus, the current review highlights the recent advances of this promising category of 1,2,3-triazole-containing hybrids incorporating diverse varieties of bioactive heterocycles such as conozole, coumarin, imidazole, benzimidazole, pyrazole, indole, oxindole, chromene, pyrane, quinazoline, chalcone, isoflavone, carbohydrates, and amides. It underlies their inhibition behavior against a wide array of infectious fungal species during 2015-2020.
Collapse
Affiliation(s)
- Michelyne Haroun
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Christophe Tratrat
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Hafedh Kochkar
- Department of Chemistry, College of Science, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Anroop B Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| |
Collapse
|
48
|
Recent studies of nitrogen containing heterocyclic compounds as novel antiviral agents: A review. Bioorg Chem 2021; 114:105076. [PMID: 34157555 DOI: 10.1016/j.bioorg.2021.105076] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/13/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022]
Abstract
N-heterocycles are important, not only because of their abundance, but above all because of their chemical, biological and technical significance. They play an important role in biological investigation such as anticancer, antiinflammatory, antibacterial, antiviral, anti-tumor, antidiabetic, etc. In this study, we focused on examining synthesized some 5- or 6-ring N-heterocyclic compounds that showed the antiviral activity in last 5 years, and investigation of these compounds structure-activity relationship studies. This review will be useful to scientists in research fields of organic synthesis, medicinal chemistry, and pharmacology.
Collapse
|
49
|
Ganesan MS, Raja KK, Murugesan S, Karankumar B, Faheem F, Thirunavukkarasu S, Shetye G, Ma R, Franzblau SG, Wan B, Rajagopal G. Quinoline‐Proline, Triazole Hybrids: Design, Synthesis, Antituberculosis, Molecular Docking, and ADMET Studies. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
| | | | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy Birla Institute of Technology and Science, Pilani Campus Pilani, Rajasthan India
| | - Banoth Karankumar
- Medicinal Chemistry Research Laboratory, Department of Pharmacy Birla Institute of Technology and Science, Pilani Campus Pilani, Rajasthan India
| | - Faheem Faheem
- Medicinal Chemistry Research Laboratory, Department of Pharmacy Birla Institute of Technology and Science, Pilani Campus Pilani, Rajasthan India
| | | | - Gauri Shetye
- Institute for Tuberculosis Research, College of Pharmacy University of Illinois at Chicago Chicago Illinois USA
| | - Rui Ma
- Institute for Tuberculosis Research, College of Pharmacy University of Illinois at Chicago Chicago Illinois USA
| | - Scott G. Franzblau
- Institute for Tuberculosis Research, College of Pharmacy University of Illinois at Chicago Chicago Illinois USA
| | - Baojie Wan
- Institute for Tuberculosis Research, College of Pharmacy University of Illinois at Chicago Chicago Illinois USA
| | - Gurusamy Rajagopal
- PG & Research Department of Chemistry Chikkanna Government Arts College Tiruppur India
| |
Collapse
|
50
|
Meuser ME, Reddy PAN, Dick A, Maurancy JM, Salvino JM, Cocklin S. Rapid Optimization of the Metabolic Stability of a Human Immunodeficiency Virus Type-1 Capsid Inhibitor Using a Multistep Computational Workflow. J Med Chem 2021; 64:3747-3766. [PMID: 33750123 DOI: 10.1021/acs.jmedchem.0c01810] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Poor metabolic stability of the human immunodeficiency virus type-1 (HIV-1) capsid (CA) inhibitor PF-74 is a major concern in its development toward clinical use. To improve on the metabolic stability, we employed a novel multistep computationally driven workflow, which facilitated the rapid design of improved PF-74 analogs in an efficient manner. Using this workflow, we designed three compounds that interact specifically with the CA interprotomer pocket, inhibit HIV-1 infection, and demonstrate enantiomeric preference. Moreover, using this workflow, we were able to increase the metabolic stability 204-fold in comparison to PF-74 in only three analog steps. These results demonstrate our ability to rapidly design CA compounds using a novel computational workflow that has improved metabolic stability over the parental compound. This workflow can be further applied to the redesign of PF-74 and other promising inhibitors with a stability shortfall.
Collapse
Affiliation(s)
- Megan E Meuser
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, Pennsylvania 19102, United States
| | - Poli Adi Narayana Reddy
- The Wistar Cancer Center Molecular Screening, The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104, United States
| | - Alexej Dick
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, Pennsylvania 19102, United States
| | - Jean Marc Maurancy
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, Pennsylvania 19102, United States
| | - Joseph M Salvino
- The Wistar Cancer Center Molecular Screening, The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104, United States
| | - Simon Cocklin
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, Pennsylvania 19102, United States
| |
Collapse
|