1
|
Rani A, Khan J, Aslam M, Ali A, Kamthan M, Pandey G, Nand B. Design, synthesis, and biological evaluation of Schiff-Base Isoxazole hybrids: Exploring novel antimicrobial agents. Bioorg Chem 2025; 159:108428. [PMID: 40184667 DOI: 10.1016/j.bioorg.2025.108428] [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: 02/20/2025] [Revised: 03/21/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025]
Abstract
The rise of antimicrobial resistance necessitates the development of novel therapeutic agents. This study presents the design, in silico studies, synthesis, and biological evaluation of Schiff-base isoxazole hybrids as potential antimicrobial agents. Computational approaches, including molecular docking, molecular dynamics (MD) simulations, density functional theory (DFT) calculations, and ADMET predictions, guided the design and assessment of these compounds. DFT analysis identified compound A20 (-0.0919 Hartree) as having the lowest energy gap, suggesting high reactivity. Molecular docking revealed strong binding affinities (-11.3 to -7.00 kcal/mol) comparable to standard antimicrobial drugs, while MD simulations confirmed the stability of protein-ligand interactions. ADMET analysis predicted favorable pharmacokinetic properties. Biological evaluations revealed promising antimicrobial potential of the synthesized compounds. A20 showed significant antifungal activity (MIC: 64 μg/mL), while A7 exhibited notable antibacterial activity (MIC: 1024 μg/mL). Further A20 in combination with standard antifungal drug fluconazole is exhibiting outstanding antifungal potential. Leading towards the possibility of developing a new drug against resistant microbial strains through combination therapy.
Collapse
Affiliation(s)
- Anjali Rani
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, Delhi, India; Department of Chemistry, SRM Institute of Science and Technology, Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, India
| | - Javed Khan
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, Delhi, India; Department of Chemistry, SRM Institute of Science and Technology, Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, India
| | - Mohd Aslam
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, Delhi, India; Department of Chemistry, SRM Institute of Science and Technology, Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, India
| | - Asghar Ali
- Clinical Biochemistry Lab, Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi -110062, India
| | - Mohan Kamthan
- Clinical Biochemistry Lab, Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi -110062, India
| | - Garima Pandey
- Department of Chemistry, SRM Institute of Science and Technology, Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, India
| | - Bhaskara Nand
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, Delhi, India.
| |
Collapse
|
2
|
Qurban J, Alqarni SA, Alalawy AI, Alshammari NAH, Ashour GRS, Alnoman MM, Katuah HA, El-Metwaly NM. Antimicrobial activity, synthesis, and docking study of some novel arylazo-1,3-thiazolopyrimidine and arylazo-1,3-thiazolopyridopyrimidine derivatives. BMC Chem 2025; 19:148. [PMID: 40437521 PMCID: PMC12121148 DOI: 10.1186/s13065-025-01506-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 05/12/2025] [Indexed: 06/01/2025] Open
Abstract
In this study, a new series of aryl azo thiazolopyrimidine and thiazolopyridopyrimidine derivatives was synthesized using novel 6-aryl-4-(2,3,6,7-tetrahydro-1H,5H-pyrido[3,2,1-ij]quinolin-9-yl)-3,4-dihydropyrimidine-2(1H)-thione and 5-aryl-7-(2,3,6,7-tetrahydro-1H,5H-pyrido[3,2,1-ij]quinolin-9-yl)-2-thioxo-2,3-dihydropyrido[2,3-d]pyrimidin-4(1H)-one scaffolds as key intermediates. Structural elucidation of all intermediates and final products was performed via IR, UV, 1H/13C-NMR, and mass spectrometry. Among the forty synthesized compounds, several exhibited significant in vitro antimicrobial activities, particularly derivatives 11a, 11b, 7a, and 7b, with potent inhibition against S. aureus, E. coli, and C. albicans. Molecular docking studies using the bacterial DNA gyrase B subunit (Protein Data Bank (PDB): 1aj6) revealed favorable binding interactions, especially for 11b, which demonstrated the best docking score and strong π-H interactions. Furthermore, DFT-based molecular modeling confirmed the stability and high electronic reactivity of selected bioactive compounds, with low HOMO-LUMO energy gaps and favorable electrostatic potential profiles. Structure-activity relationship (SAR) analysis indicated that electronic effects, lipophilicity, and heteroaromatic substitution patterns critically influence antimicrobial potency. These findings support the potential of thiazolopyridopyrimidine derivatives as promising scaffolds for future antimicrobial drug development.
Collapse
Affiliation(s)
- Jihan Qurban
- Department of Chemistry, College of Science, Umm Al-Qura University, 24230, Makkah, Saudi Arabia
| | - Sara A Alqarni
- Department of Chemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Adel I Alalawy
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Nawaa Ali H Alshammari
- Department of Chemistry, College of Science, Northern Border University, 73222, Arar, Saudi Arabia
| | - Gadeer R S Ashour
- Department of Chemistry, College of Science, Umm Al-Qura University, 24230, Makkah, Saudi Arabia
| | - Maryam M Alnoman
- Biology Department, Faculty of Science, Taibah University, Yanbu, Saudi Arabia
| | - Hanadi A Katuah
- Department of Chemistry, College of Science, Umm Al-Qura University, 24230, Makkah, Saudi Arabia
| | - Nashwa M El-Metwaly
- Department of Chemistry, College of Science, Umm Al-Qura University, 24230, Makkah, Saudi Arabia.
- Department of Chemistry, Faculty of Science, Mansoura University, El-Gomhoria Street 35516, Mansoura, Egypt.
| |
Collapse
|
3
|
Xu Z, Liu J, Zhuang Y. The anti-Acinetobacter baumannii therapeutic potential of azole hybrids: A mini-review. Arch Pharm (Weinheim) 2025; 358:e2400592. [PMID: 40040257 DOI: 10.1002/ardp.202400592] [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: 07/19/2024] [Revised: 12/19/2024] [Accepted: 02/11/2025] [Indexed: 03/06/2025]
Abstract
Acinetobacter baumannii is one of the major causes of severe hospital- and community-acquired infections, posing a significant threat to human lives. A. baumannii has already generated resistance to almost all of the currently available antibiotics, but no new class of antibacterials have been launched for the treatment of infections caused by A. baumannii in the last half century, creating an urgent need to develop novel antibacterials. Azoles as a broad class of five-membered nitrogen-containing aromatic heterocycles are privileged pharmacophores widely found in pharmaceuticals. Azoles could target on diverse enzymes, proteins, and receptors in A. baumannii via various noncovalent interactions. Particularly, azole hybrids have potential advantages in increasing therapeutic efficacy and circumventing drug resistance, representing useful scaffolds for the discovery of novel anti-A. baumannii agents. This review outlines the current scenario of the antibacterial therapeutic potential of azole hybrids against A. baumannii, developed from 2020 onwards, aiming to provide potential candidates for further preclinical/clinical evaluations and facilitate the rational design of more effective candidates.
Collapse
Affiliation(s)
- Zhi Xu
- Huanghuai University Industry Innovation & Research and Development Institute, Huanghuai University, Zhumadian, People's Republic of China
| | - Junna Liu
- Huanghuai University Industry Innovation & Research and Development Institute, Huanghuai University, Zhumadian, People's Republic of China
| | - Yafei Zhuang
- Huanghuai University Industry Innovation & Research and Development Institute, Huanghuai University, Zhumadian, People's Republic of China
| |
Collapse
|
4
|
Shaheen MA, Darwish KM, Kishk SM, El-Sayed MAA, Salama I. Development of 1,2,3-triazole hybrids as multi-faced anticancer agents co-targeting EGFR/mTOR pathway and tubulin depolymerization. Bioorg Chem 2025; 156:108153. [PMID: 39855112 DOI: 10.1016/j.bioorg.2025.108153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025]
Abstract
Novel 1,2,3-triazole hybrids bearing various substituents have been synthesized as potential anticancer agents. Ligand-based approach has been adopted to design these compounds relying on the hybridization of 1,2,3-triazole with α,β-unsaturated carbonyl, 5- and 6-membered heterocyclic scaffolds. All synthesized members were investigated for their cytotoxic potency against nine types comprising 60 panels of human cancerous cells by the US National Cancer Institute: Development Therapeutic Program (US_NCI_DTP). Among the tested members, 4b, 4e, and 4h showed prominent cytotoxic effects (> 80 % growth inhibition: GI) on a wide panel of tested cancer cell lines, mainly melanoma and colorectal cancer redeeming their selection for five dose testing. Presenting low nanomolar GI50 concentrations, two representative potent anticancer compounds 4b and 4e were subjected to cytotoxicity testing on colon normal cell (FHC) to investigate their safety window and they showed less toxicity to normal cells at the concentration required to produce anticancer effect. Furthermore, 4b and 4e were exposed to additional mechanistic studies in colorectal cancer cell HCT-116 suggesting multifaceted mechanisms of action. A study into the effects of cytotoxic chemicals 4b and 4e on cell cycle progression regulation showed triggered the arrest of cell cycles during the G1 and S phases. Moreover, 4b and 4e caused cell death mainly through apoptosis the thing that has been reinforced by the elevated Bax: Bcl2 ratio, as well as concentrations of caspases 3 and 9 within HCT-116. Further, both compounds showed prominent inhibition profiles against tubulin polymerization as well as EGFR catalytic activity reaching down to low-digit micromolar and sub-micromolar concentrations, respectively, as compared to positive reference controls. Compounds' impacts on gene expression of cancer-associated and EGFR-downstream signaling markers including TNFα, IL-6, and mTOR, were explored in HCT-116 highlighted significant downregulations versus the untreated cells. Docking studies demonstrated the specific fit of 4b and 4e into EGFR and the colchicine binding site of tubulin.
Collapse
Affiliation(s)
- Mennatallah A Shaheen
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522 Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University, Egypt.
| | - Khaled M Darwish
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522 Egypt; Medicinal Chemistry Department, Faculty of Pharmacy, Galala University, New Galala 43511 Egypt.
| | - Safaa M Kishk
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522 Egypt.
| | - Magda A-A El-Sayed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University, Egypt; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Ismail Salama
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522 Egypt.
| |
Collapse
|
5
|
Jeon H, Kim YG, Lee JH, Lee J. Antibiofilm Activities of Halogenated Pyrimidines Against Enterohemorrhagic Escherichia coli O157:H7. Int J Mol Sci 2025; 26:1386. [PMID: 39941153 PMCID: PMC11818689 DOI: 10.3390/ijms26031386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) is a significant public health concern due to its ability to form biofilms, enhancing its resistance to antimicrobials and contributing to its persistence in food processing environments. Traditional antibiotics often fail to target these biofilms effectively, leading to increased bacterial resistance. This study aims to explore the efficacy of novel antibiofilm agents, specifically halogenated pyrimidine derivatives, against EHEC. We screened pyrimidine and 31 halogenated pyrimidine derivatives for their antimicrobial and antibiofilm activities against EHEC using biofilm quantification assays, SEM analysis, motility, and curli production assessments. Our findings reveal that certain halogenated pyrimidine derivatives, notably 2-amino-5-bromopyrimidine (2A5BP), 2-amino-4-chloropyrrolo[2,3-d]pyrimidine (2A4CPP), and 2,4-dichloro-5-iodo-7H-pyrrolo[2,3-d]pyrimidine (2,4DC5IPP) at 50 µg/mL, exhibited significant inhibitory effects on EHEC biofilm formation without affecting bacterial growth, suggesting a targeted antibiofilm action. These compounds effectively reduced curli production and EHEC motility, essential factors for biofilm integrity and development. qRT-PCR analysis revealed that two active compounds downregulated the expression of key curli genes (csgA and csgB), leading to reduced bacterial adhesion and biofilm formation. Additionally, in silico ADME-Tox profiles indicated that these compounds exhibit favorable drug-like properties and lower toxicity compared with traditional pyrimidine. This study highlights the potential of halogenated pyrimidine derivatives as effective antibiofilm agents against EHEC, offering a promising strategy for enhancing food safety and controlling EHEC infections. The distinct mechanisms of action of these compounds, particularly in inhibiting biofilm formation and virulence factors without promoting bacterial resistance, underscore their therapeutic potential.
Collapse
Affiliation(s)
| | | | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea; (H.J.); (Y.-G.K.)
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea; (H.J.); (Y.-G.K.)
| |
Collapse
|
6
|
Zhou M, Duan X, Jin T, Feng X, Liu Y, Wang S, Feng J, Zhang M, Chai T, Mao B, Shao S, Jin G. Design, synthesis, and antitumor activity evaluation of BF 3-o, m, p-phenylenediamine bridged with pyrimidine-indole BF 3 adduction derivatives. Mol Divers 2025; 29:425-437. [PMID: 39030284 DOI: 10.1007/s11030-024-10863-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/26/2024] [Indexed: 07/21/2024]
Abstract
Fluorescent drugs and pyrimidine-indole scaffolds have been shown to have advantages in cancer treatment. Fluorescent antitumor drugs BF3-o, m, p-phenylenediamine pyrimidine-indole derivatives (PYB1, PYB2, and PYB3) were synthesized by linking pyrimidine and indole groups with aniline through a simple step and introducing BF3. The drugs exhibit promising antitumor activity and their fluorescent properties make them useful for imaging purposes. The optical properties of the three compounds have been investigated. All of them have fluorescent properties and compound PYB2 has good fluorescent properties. Additionally, the in vitro cytotoxicity of these compounds was evaluated against the human cancer cell line HeLa and the human normal cell line L02. The inhibition rates of HeLa cells treated with PYB1, PYB2, and PYB3 at a concentration of 19.2 μg/mL were 80.91%, 77.72%, and 65.94%, respectively. These results indicate a strong inhibitory effect on cancer cells. Further through the cell imaging experiment, we can see that PYB2 can enter the cell through the cell membrane through the fluorescence scattering diagram, which has good biocompatibility. In addition, the possible interactions between the compounds and Ras protein active sites were analyzed by molecular docking. The three compounds can bind well to Ras protein through hydrogen bonding. This study provides a basis for the development and modification of pyrimidine-indole fluorescent anticancer drugs. Compound PYB2 shows potential as a fluorescent anticancer drug.
Collapse
Affiliation(s)
- Meng Zhou
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Xiujie Duan
- Zhenjiang Hospital Affiliated to Jiangsu University, Zhenjiang, 212013, China
| | - Tao Jin
- Yixing Hospital Affiliated to Jiangsu University, Yixing, 214200, China
| | - Xibing Feng
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Ying Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Shuo Wang
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Jiankang Feng
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Mengtong Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Tiantian Chai
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Boneng Mao
- Yixing Hospital Affiliated to Jiangsu University, Yixing, 214200, China.
| | - Shihe Shao
- Yixing Hospital Affiliated to Jiangsu University, Yixing, 214200, China.
| | - Guofan Jin
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
7
|
Dai J, Li Q, Li Z, Zang Z, Luo Y, Zhou C. Discovery of Quinazolone Pyridiniums as Potential Broad-Spectrum Antibacterial Agents. Molecules 2025; 30:243. [PMID: 39860113 PMCID: PMC11767251 DOI: 10.3390/molecules30020243] [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: 11/29/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
The overprescription of antibiotics in medicine and agriculture has accelerated the development and spread of antibiotic resistance in bacteria, which severely limits the arsenal available to clinicians for treating bacterial infections. This work discovered a new class of heteroarylcyanovinyl quinazolones and quinazolone pyridiniums to surmount the increasingly severe bacterial resistance. Bioactive assays manifested that the highly active compound 19a exhibited strong inhibition against MRSA and Escherichia coli with extremely low MICs of 0.5 μg/mL, being eightfold more active than that of norfloxacin (MICs = 4 μg/mL). The highly active 19a with rapid bactericidal properties displayed imperceptible resistance development trends, negligible hemolytic toxicity, and effective biofilm inhibitory effects. Preliminary explorations on antibacterial mechanisms revealed that compound 19a could cause membrane damage, embed in intracellular DNA to hinder bacterial DNA replication, and induce metabolic dysfunction. Surprisingly, active 19a was found to trigger the conformational change in PBP2a of MRSA to open the active site, which might account for its high inhibition against MRSA. In addition, the little effect of molecule 19a on the production of reactive oxygen species indicated that bacterial death was not caused by oxidative stress. The above comprehensive analyses highlighted the large potential of quinazolone pyridiniums as multitargeting broad-spectrum antibacterial agents.
Collapse
Affiliation(s)
- Jie Dai
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Qianyue Li
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Ziyi Li
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Zhonglin Zang
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Yan Luo
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Chenghe Zhou
- Institute of Bioorganic & Medicinal Chemistry, Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| |
Collapse
|
8
|
El-Gharably AA, Nassar AA, El-Ganzory NM, Saad-Allah KM, El-Barbary AA. Sulfoxidation of pyrimidine thioate derivatives and study their biological activities. Sci Rep 2025; 15:1024. [PMID: 39762326 PMCID: PMC11704135 DOI: 10.1038/s41598-024-83050-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
In a quest to innovate biologically active molecules, the benzoylation of 4,6-dimethylpyrimidine-2-thiol hydrochloride (1) with benzoyl chloride derivatives was employed to produce a series of pyrimidine benzothioate derivatives (2-5). Subsequent sulfoxidation of these derivatives (2-5) using hydrogen peroxide and glacial acetic acid yielded a diverse array of pyrimidine sulfonyl methanone derivatives (6-9). In parallel, the sulfoxidation of pyrimidine sulfonothioates (10-12) yielded sulfonyl sulfonyl pyrimidines (13-15), originating from the condensation of compound 1 with sulfonyl chloride derivatives. The newly synthesized compounds underwent characterization via FT-IR, NMR, mass spectrometry, and elemental analyses. Biological screenings unveiled interesting properties: compounds 1 and 6 exhibited significant antimicrobial potency against S. epidermidis and S. haemolyticus, whereas compound 11 showed distinct insensitivity. Excitingly, compounds 12 and 6 showcased robust antioxidant activity by efficiently scavenging DPPH• radical, underscoring their potential in oxidative stress mitigation. Notably, compounds 10 and 12 displayed promising anti-tumor effects, with compound 12 demonstrating superior efficacy against the MCF-7 breast cancer cell line compared to compound 10. The study revealed a spectrum of biological activities across the synthesized derivatives, with modifications often resulting in diminished bioactivity compared to the parent compound 1. These findings shed light on the intricate relationship between chemical modifications and biological properties, offering valuable insights for future drug discovery endeavors.
Collapse
Affiliation(s)
- Atif A El-Gharably
- Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - A A Nassar
- Chemistry Department, Faculty of Science, Menoufia University, Shibin El-Kom, 32511, Egypt
| | - N M El-Ganzory
- Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Khalil M Saad-Allah
- Botany Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - A A El-Barbary
- Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
9
|
Sim M, Kim YG, Lee JH, Lee J. Antibiofilm Activities of Multiple Halogenated Pyrimidines Against Staphylococcus aureus. Int J Mol Sci 2024; 25:12830. [PMID: 39684543 DOI: 10.3390/ijms252312830] [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: 11/01/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Staphylococcus aureus, prevalent in hospital and community settings, forms biofilms that are highly resistant to antibiotics and immune responses, complicating treatment and contributing to chronic infections. These challenges underscore the need for novel treatments that target biofilm formation and effectively reduce bacterial virulence. This study investigates the antibiofilm and antimicrobial efficacy of novel halogenated pyrimidine derivatives against S. aureus, focusing on three compounds identified as potent biofilm inhibitors: 2,4-dichloro-5-fluoropyrimidine (24DC5FP), 5-bromo-2,4-dichloro-7H-pyrrolo[2,3-d]pyrimidine (24DC5BPP), and 2,4-dichloro-5-iodo-7H-pyrrolo[2,3-d]pyrimidine (24DC5IPP). The three active compounds are bacteriostatic. In particular, 24DC5FP at 5 µg/mL achieved a 95% reduction in hemolysis with a minimum inhibitory concentration (MIC) of 50 µg/mL. Interestingly, 24DC5FP increased cell size and produced wrinkled colonies. qRT-PCR analysis showed that 24DC5FP suppressed the gene expressions of agrA and RNAIII (quorum sensing regulator and effector), hla (α-hemolysin), nuc1 (nucleases nuc1), and saeR (S. aureus virulence regulator). These findings suggest that extensive halogenation enhances the antibiofilm and antivirulence activities of pyrimidine derivatives, offering a promising strategy for combatting S. aureus infections, including those resistant to conventional treatments.
Collapse
Affiliation(s)
- MinHwi Sim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
10
|
Chen YW. Current scenario of indole hybrids with antibacterial potential against Acinetobacter baumannii pathogens: A mini-review. Arch Pharm (Weinheim) 2024; 357:e2400440. [PMID: 38986447 DOI: 10.1002/ardp.202400440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Abstract
Acinetobacter baumannii with the capability to "escape" almost all currently available antibacterials is eroding the safety of basic medical interventions and is an increasing cause of mortality globally, prompting a substantial requirement for new classes of antibacterial agents. Indoles participate in the regulation of persistent bacterial formation, biofilm formation, plasmid stability, and drug resistance. In particular, indole hybrids demonstrated promising antibacterial activity against both drug-sensitive and drug-resistant A. baumannii pathogens, representing a fertile source for the discovery of novel therapeutic agents for clinical deployment in controlling A. baumannii infections. This mini-review outlines the current innovations of indole hybrids with antibacterial activity against A. baumannii pathogens, covering articles published from 2020 to the present, to open new avenues for exploring novel anti-A. baumannii candidates.
Collapse
Affiliation(s)
- Yi-Wen Chen
- Department of Internal Medicine, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
11
|
Chen M, Cheng S, Dai X, Yu J, Wang H, Xu B, Luo H, Xu G. Design, Synthesis, and Biological Evaluation of Novel Quinazoline Derivatives Possessing a Trifluoromethyl Moiety as Potential Antitumor Agents. Chem Biodivers 2024; 21:e202301776. [PMID: 38602834 DOI: 10.1002/cbdv.202301776] [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: 11/09/2023] [Accepted: 03/13/2024] [Indexed: 04/13/2024]
Abstract
A novel series of trifluoromethyl-containing quinazoline derivatives with a variety of functional groups was designed, synthesized, and tested for their antitumor activity by following a pharmacophore hybridization strategy. Most of the 20 compounds displayed moderate to excellent antiproliferative activity against five different cell lines (PC3, LNCaP, K562, HeLa, and A549). After three rounds of screening and structural optimization, compound 10 b was identified as the most potent one, with IC50 values of 3.02, 3.45, and 3.98 μM against PC3, LNCaP, and K562 cells, respectively, which were comparable to the effect of the positive control gefitinib. To further explore the mechanism of action of 10 b against cancer, experiments focusing on apoptosis induction, cell cycle arrest, and cell migration assay were conducted. The results showed that 10 b was able to induce apoptosis and prevent tumor cell migration, but had no effect on the cell cycle of tumor cells.
Collapse
Affiliation(s)
- Mingxiu Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Xing Dai
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 561113, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - HuiDi Wang
- The Affiliated Hospital of Wuhan Sports University, Wuhan, 430079, China
| | - BiXue Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Guangcan Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| |
Collapse
|
12
|
Tiwari G, Mishra VK, Khanna A, Tyagi R, Sagar R. Synthesis of Chirally Enriched Pyrazolylpyrimidinone-Based Glycohybrids via Annulation of Glycals with 2-Hydrazineylpyrimidin-4(3 H)-ones. J Org Chem 2024; 89:5000-5009. [PMID: 38471017 DOI: 10.1021/acs.joc.4c00211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
A new strategy for synthesizing chirally enriched pyrazolylpyrimidinone-based glycohybrids has been achieved, employing an annulation approach in ethanol without any additives or catalysts under microwave conditions. The designed compounds were obtained within a short reaction time (5 min). This method offers several advantages, including mild reaction conditions, a green solvent, and a metal-free approach. Furthermore, the protocol demonstrated a broad substrate scope, successfully incorporating various functional groups with stereochemical diversity and furnishing chirally enriched molecules.
Collapse
Affiliation(s)
- Ghanshyam Tiwari
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Vinay Kumar Mishra
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Ashish Khanna
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Rajdeep Tyagi
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ram Sagar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
13
|
Upadhyay DB, Mokariya JA, Patel PJ, Patel SG, Das A, Nandi A, Nogales J, More N, Kumar A, Rajani DP, Narayan M, Kumar J, Banerjee S, Sahoo SK, Patel HM. Indole clubbed 2,4-thiazolidinedione linked 1,2,3-triazole as a potent antimalarial and antibacterial agent against drug-resistant strain and molecular modeling studies. Arch Pharm (Weinheim) 2024; 357:e2300673. [PMID: 38247229 DOI: 10.1002/ardp.202300673] [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: 11/19/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
In the face of escalating challenges of microbial resistance strains, this study describes the design and synthesis of 5-({1-[(1H-1,2,3-triazol-4-yl)methyl]-1H-indol-3-yl}methylene)thiazolidine-2,4-dione derivatives, which have demonstrated significant antimicrobial properties. Compared with the minimum inhibitory concentrations (MIC) values of ciprofloxacin on the respective strains, compounds 5a, 5d, 5g, 5l, and 5m exhibited potent antibacterial activity with MIC values ranging from 16 to 25 µM. Almost all the synthesized compounds showed lower MIC compared to standards against vancomycin-resistant enterococcus and methicillin-resistant Staphylococcus aureus strains. Additionally, the majority of the synthesized compounds demonstrated remarkable antifungal activity, against Candida albicans and Aspergillus niger, as compared to nystatin, griseofulvin, and fluconazole. Furthermore, the majority of compounds exhibited notable inhibitory effects against the Plasmodium falciparum strain, having IC50 values ranging from 1.31 to 2.79 μM as compared to standard quinine (2.71 μM). Cytotoxicity evaluation of compounds 5a-q on SHSY-5Y cells at up to 100 μg/mL showed no adverse effects. Comparison with control groups highlights their noncytotoxic characteristics. Molecular docking confirmed compound binding to target active sites, with stable protein-ligand complexes displaying drug-like molecules. Molecular dynamics simulations revealed dynamic stability and interactions. Rigorous tests and molecular modeling unveil the effectiveness of the compounds against drug-resistant microbes, providing hope for new antimicrobial compounds with potential safety.
Collapse
Affiliation(s)
- Dipti B Upadhyay
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Jaydeep A Mokariya
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Paras J Patel
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Subham G Patel
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Anwesha Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India
| | - Arijit Nandi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Joaquina Nogales
- Department of Cellular and Systems Medicine, University of Dundee, Dundee, UK
| | - Nachiket More
- School of Chemistry, University of St. Andrews, St. Andrews, UK
| | - Amit Kumar
- School of Chemistry, University of St. Andrews, St. Andrews, UK
| | - Dhanji P Rajani
- Microcare Laboratory and Tuberculosis Diagnosis & Research Center, Surat, Gujarat, India
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, The University of Texas at El Paso, El Paso, Texas, USA
| | - Jyotish Kumar
- Department of Chemistry and Biochemistry, The University of Texas at El Paso, El Paso, Texas, USA
| | - Sourav Banerjee
- Department of Cellular and Systems Medicine, University of Dundee, Dundee, UK
| | - Suban K Sahoo
- Department of Chemistry, SV National Institute of Technology, Surat, Gujarat, India
| | - Hitendra M Patel
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| |
Collapse
|
14
|
Nammalwar B, Bunce RA. Recent Advances in Pyrimidine-Based Drugs. Pharmaceuticals (Basel) 2024; 17:104. [PMID: 38256937 PMCID: PMC10820437 DOI: 10.3390/ph17010104] [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: 11/27/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 01/24/2024] Open
Abstract
Pyrimidines have become an increasingly important core structure in many drug molecules over the past 60 years. This article surveys recent areas in which pyrimidines have had a major impact in drug discovery therapeutics, including anti-infectives, anticancer, immunology, immuno-oncology, neurological disorders, chronic pain, and diabetes mellitus. The article presents the synthesis of the medicinal agents and highlights the role of the biological target with respect to the disease model. Additionally, the biological potency, ADME properties and pharmacokinetics/pharmacodynamics (if available) are discussed. This survey attempts to demonstrate the versatility of pyrimidine-based drugs, not only for their potency and affinity but also for the improved medicinal chemistry properties of pyrimidine as a bioisostere for phenyl and other aromatic π systems. It is hoped that this article will provide insight to researchers considering the pyrimidine scaffold as a chemotype in future drug candidates in order to counteract medical conditions previously deemed untreatable.
Collapse
Affiliation(s)
- Baskar Nammalwar
- Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, CA 92121, USA;
| | - Richard A. Bunce
- Department of Chemistry, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
15
|
Li J, Gu A, Nong XM, Zhai S, Yue ZY, Li MY, Liu Y. Six-Membered Aromatic Nitrogen Heterocyclic Anti-Tumor Agents: Synthesis and Applications. CHEM REC 2023; 23:e202300293. [PMID: 38010365 DOI: 10.1002/tcr.202300293] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/19/2023] [Indexed: 11/29/2023]
Abstract
Cancer stands as a serious malady, posing substantial risks to human well-being and survival. This underscores the paramount necessity to explore and investigate novel antitumor medications. Nitrogen-containing compounds, especially those derived from natural sources, form a highly significant category of antitumor agents. Among these, antitumor agents with six-membered aromatic nitrogen heterocycles have consistently attracted the attention of chemists and pharmacologists. Accordingly, we present a comprehensive summary of synthetic strategies and clinical implications of these compounds in this review. This entails an in-depth analysis of synthesis pathways for pyridine, quinoline, pyrimidine, and quinazoline. Additionally, we explore the historical progression, targets, mechanisms of action, and clinical effectiveness of small molecule inhibitors possessing these structural features.
Collapse
Affiliation(s)
- Jiatong Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Ao Gu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Xiao-Mei Nong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Shuyang Zhai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Zhu-Ying Yue
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Meng-Yao Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Yingbin Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| |
Collapse
|
16
|
Tiwari G, Khanna A, Mishra VK, Sagar R. Recent developments on microwave-assisted organic synthesis of nitrogen- and oxygen-containing preferred heterocyclic scaffolds. RSC Adv 2023; 13:32858-32892. [PMID: 37942237 PMCID: PMC10628940 DOI: 10.1039/d3ra05986c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023] Open
Abstract
In recent decades, the utilization of microwave energy has experienced an extraordinary surge, leading to the introduction of innovative and revolutionary applications across various fields of chemistry such as medicinal chemistry, materials science, organic synthesis and heterocyclic chemistry. Herein, we provide a comprehensive literature review on the microwave-assisted organic synthesis of selected heterocycles. We highlight the use of microwave irradiation as an effective method for constructing a diverse range of molecules with high yield and selectivity. We also emphasize the impact of microwave irradiation on the efficient synthesis of N- and O-containing heterocycles that possess bioactive properties, such as anti-cancer, anti-proliferative, and anti-tumor activities. Specific attention is given to the efficient synthesis of pyrazolopyrimidines-, coumarin-, quinoline-, and isatin-based scaffolds, which have been extensively studied for their potential in drug discovery. The article provides valuable insights into the recent synthetic protocols and trends for the development of new drugs using heterocyclic molecules.
Collapse
Affiliation(s)
- Ghanshyam Tiwari
- Department of Chemistry, Institute of Science, Banaras Hindu University Varanasi 221005 India
| | - Ashish Khanna
- Department of Chemistry, Institute of Science, Banaras Hindu University Varanasi 221005 India
| | - Vinay Kumar Mishra
- Department of Chemistry, Institute of Science, Banaras Hindu University Varanasi 221005 India
| | - Ram Sagar
- Department of Chemistry, Institute of Science, Banaras Hindu University Varanasi 221005 India
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University New Delhi 110067 India
| |
Collapse
|
17
|
Nibin Joy M, Guda MR, Zyryanov GV. Evaluation of Anti-Inflammatory and Anti-Tubercular Activity of 4-Methyl-7-Substituted Coumarin Hybrids and Their Structure Activity Relationships. Pharmaceuticals (Basel) 2023; 16:1326. [PMID: 37765134 PMCID: PMC10535168 DOI: 10.3390/ph16091326] [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: 08/13/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Four sets of previously synthesized 4-methyl-7-substituted coumarin derivatives were screened for their in vitro anti-inflammatory and anti-tubercular activities. The anti-inflammatory potential of 3a-t, 5a-o, 6a-n, and 7a-f synthesized compounds was evaluated by an anti-denaturation assay using diclofenac sodium as the reference standard. Evaluation of the anti-tuberculous activity of the mentioned compounds was performed by the Resazurin test method against four different TB strains using rifampicin and isoniazid as reference drugs. Based on the anti-inflammatory results, compounds 3o, 5f, 6c, and 7d proved to be the most active compounds in their respective series. Additionally, compounds 3k-n, 5b-d, 6d-f, 6k, 7a, and 7f were found to be the most potent anti-tuberculous agents. In fact, most of the screened compounds exhibited promising activity profiles compared to the respective standard drugs. The structure-activity connections revealed a few intriguing aspects, indicating that the presence of electron-donating and nitrogen-rich fragments boost the anti-inflammatory effects of the examined compounds. However, the presence of electron-withdrawing substituents was required to boost the anti-tubercular activity of the evaluated compounds.
Collapse
Affiliation(s)
- Muthipeedika Nibin Joy
- Institute of Chemical Engineering, Ural Federal University Named after the First President of Russia B. N. Yeltsin, 28 Mira St., Yekaterinburg 620002, Russia
| | - Mallikarjuna R. Guda
- Institute of Chemical Engineering, Ural Federal University Named after the First President of Russia B. N. Yeltsin, 28 Mira St., Yekaterinburg 620002, Russia
| | - Grigory V. Zyryanov
- Institute of Chemical Engineering, Ural Federal University Named after the First President of Russia B. N. Yeltsin, 28 Mira St., Yekaterinburg 620002, Russia
- Ural Division of the Russian Academy of Sciences, I. Ya. Postovskiy Institute of Organic Synthesis, 22 S. Kovalevskoy Street, Yekaterinburg 620219, Russia
| |
Collapse
|