1
|
Zaware N, Rasal N, Lambate V, Jagtap S. Biological evaluation of newly synthesized α-benzil monoxime thiocarbohydrazide derivatives as an antimicrobial and anticancer agent: In vitro screening and ADMET predictions. Bioorg Med Chem Lett 2025; 118:130079. [PMID: 39710140 DOI: 10.1016/j.bmcl.2024.130079] [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: 06/11/2024] [Revised: 12/01/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
The current comprehensive study showcases a meticulous synthesis of novel class of α-benzilmonoxime thiocarbohydrazide (BMOTC) derivatives, and manifesting their multifaceted potential as antibacterial, antifungal, and anticancer agents. The synthesis of target compounds was performed in three phases using literature methods. In the first step, benzilmonoxime is synthesized using benzil and hydroxyl amine hydrochloride, followed by benzilmonoxime imine using thiocarbohydrazide. The final stage involves combining BMOTC imine with various aldehydes and ketones. The antibacterial and antifungal activities of the synthesized derivatives against five bacterial panels, both Gram-positive and Gram-negative, and one fungal pathogen have been screened. Twelve of the twenty-four synthetic derivatives showed noteworthy activity; eight derivatives exhibited growth inhibition (GI) >73 % against Acinetobacter baumannii, two exhibited GI >95 % against Escherichia coli, and two exhibited GI >93 % against Candida albicans at concentration 32 μg/mL. Further assessment revealed that two derivatives 5v and 5w, exhibited negligible cytotoxicity towards human embryonic kidney cells (HK-293) and human red blood cells (RBC), signifying their promising safety profile at concentration 32 μg/mL (GI against Candida albicans - 97.51 % and 93.71 % respectively). The synthesized compounds were subjected to in vitro cytostatic activity, where a rigorous scrutiny against a diverse panel of NCI 60 cancer cell lines representing various malignancies was carried out. A total of eleven compounds emerged as promising candidates, demonstrating significant growth percent (GP) at a concentration of 10 µM. Notably, compounds 5d, 5h, and 5x, turned up as standout performers, exhibiting potent anticancer activity across multiple cancer types, including colon, CNS, melanoma, and breast cancers. Of particular interest, compound 5d displayed notable antiproliferative effects against leukemia cancer cell lines RPMI-8226 & SR, while maintaining non-cytotoxicity against the same. Compound 5h showcased activity against ovarian, non-small cell lung, and prostate cancers, without inducing cytotoxic effects. Compound 5x demonstrated remarkable anticancer activity against leukemia and breast cancer cell lines, further bolstered by its non-cytotoxic nature. A compelling aspect of this study is the comparative analysis with the established drug molecule sunitinib, revealing that compounds 5d, 5h, and 5x exhibit superior potency. These findings not only highlight the therapeutic potential of the BMOTC derivatives but also underscore their viability as promising candidates for future drug development endeavours. This study serves as a pivotal step towards harnessing the untapped therapeutic potential of BMOTC derivatives in combating microbial infections and advancing cancer therapy.
Collapse
Affiliation(s)
- Navnath Zaware
- Department of Chemistry, PDEA's Baburaoji Gholap College, Sangvi, Pune 27, India
| | - Nishant Rasal
- Department of Chemistry, PDEA's Baburaoji Gholap College, Sangvi, Pune 27, India
| | - Vinayak Lambate
- Department of Chemistry, PDEA's Baburaoji Gholap College, Sangvi, Pune 27, India
| | - Sangeeta Jagtap
- Department of Chemistry, PDEA's Baburaoji Gholap College, Sangvi, Pune 27, India.
| |
Collapse
|
2
|
Peretz E, Musa S. Design, Synthesis, and Characterization of Novel Cannabidiol-Based Derivatives with Potent Antioxidant Activities. Int J Mol Sci 2024; 25:9579. [PMID: 39273525 PMCID: PMC11395037 DOI: 10.3390/ijms25179579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
In recent years, extensive research has focused on cannabidiol (CBD), a well-studied non-psychoactive component of the plant-derived cannabinoids. CBD has shown significant therapeutic potential for treating various diseases and disorders, including antioxidants and anti-inflammatory effects. Due to the promising therapeutic effect of CBD in a wide variety of diseases, synthetic derivatization of this compound has attracted the attention of drug discovery in both industry and academia. In the current research, we focused on the derivatization of CBD by introducing Schiff base moieties, particularly (thio)-semicarbazide and aminoguanidine motifs, at the 3-position of the olivetolic ring. We have designed, synthesized, and characterized new derivatives based on CBD's framework, specifically aminoguanylhydrazone- and (thio)-semicarbazones-CBD-aldehyde compounds. Their antioxidant potential was assessed using FRAP and DPPH assays, alongside an evaluation of their effect on LDL oxidation induced by Cu2+ and AAPH. Our findings suggest that incorporating the thiosemicarbazide motif into the CBD framework produces a potent antioxidant, warranting further investigation.
Collapse
Affiliation(s)
- Eliav Peretz
- Department of Biotechnology, Tel-Hai Academic College, Kiryat Shmona 11016, Israel
- Natural Compounds and Organic Synthesis Laboratory, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
| | - Sanaa Musa
- Department of Biotechnology, Tel-Hai Academic College, Kiryat Shmona 11016, Israel
- Natural Compounds and Organic Synthesis Laboratory, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
| |
Collapse
|
3
|
Yang K, Yin D, Sun Y, Yang Z, Li Y, Xu L, Du Y. Synthesis of Fluoromethylated Chromones and Their Heteroatom Analogues via Sodium Fluoromethanesulfinate-Enabled Direct Fluoromethylation. J Org Chem 2024; 89:565-575. [PMID: 38115769 DOI: 10.1021/acs.joc.3c02301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
An array of biologically interesting tri/difluoromethylated chromones and their heteroatom analogues were conveniently synthesized from the reaction of chromones and their heteroatom analogues with CF3SO2Na or HCF2SO2Na in the presence of tert-butyl hydroperoxide under mild conditions. A mechanistic pathway involving the generation of the electrophilic tri/difluoromethyl radical, followed with the radical substitution of chromones and their heteroatom analogues, was postulated.
Collapse
Affiliation(s)
- Kaiyue Yang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Dongxue Yin
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yuli Sun
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Zhifang Yang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yadong Li
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Lingzhi Xu
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yunfei Du
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
4
|
Murthy S, Nizi MG, Maksimainen MM, Massari S, Alaviuhkola J, Lippok BE, Vagaggini C, Sowa ST, Galera-Prat A, Ashok Y, Venkannagari H, Prunskaite-Hyyryläinen R, Dreassi E, Lüscher B, Korn P, Tabarrini O, Lehtiö L. [1,2,4]Triazolo[3,4- b]benzothiazole Scaffold as Versatile Nicotinamide Mimic Allowing Nanomolar Inhibition of Different PARP Enzymes. J Med Chem 2023; 66:1301-1320. [PMID: 36598465 PMCID: PMC9884089 DOI: 10.1021/acs.jmedchem.2c01460] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We report [1,2,4]triazolo[3,4-b]benzothiazole (TBT) as a new inhibitor scaffold, which competes with nicotinamide in the binding pocket of human poly- and mono-ADP-ribosylating enzymes. The binding mode was studied through analogues and cocrystal structures with TNKS2, PARP2, PARP14, and PARP15. Based on the substitution pattern, we were able to identify 3-amino derivatives 21 (OUL243) and 27 (OUL232) as inhibitors of mono-ARTs PARP7, PARP10, PARP11, PARP12, PARP14, and PARP15 at nM potencies, with 27 being the most potent PARP10 inhibitor described to date (IC50 of 7.8 nM) and the first PARP12 inhibitor ever reported. On the contrary, hydroxy derivative 16 (OUL245) inhibits poly-ARTs with a selectivity toward PARP2. The scaffold does not possess inherent cell toxicity, and the inhibitors can enter cells and engage with the target protein. This, together with favorable ADME properties, demonstrates the potential of TBT scaffold for future drug development efforts toward selective inhibitors against specific enzymes.
Collapse
Affiliation(s)
- Sudarshan Murthy
- Faculty
of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu90220, Finland
| | - Maria Giulia Nizi
- Department
of Pharmaceutical Sciences, University of
Perugia, Perugia06123, Italy
| | - Mirko M. Maksimainen
- Faculty
of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu90220, Finland
| | - Serena Massari
- Department
of Pharmaceutical Sciences, University of
Perugia, Perugia06123, Italy
| | - Juho Alaviuhkola
- Faculty
of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu90220, Finland
| | - Barbara E. Lippok
- Institute
of Biochemistry and Molecular Biology, RWTH
Aachen University, Aachen52074, Germany
| | - Chiara Vagaggini
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, SienaI-53100, Italy
| | - Sven T. Sowa
- Faculty
of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu90220, Finland
| | - Albert Galera-Prat
- Faculty
of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu90220, Finland
| | - Yashwanth Ashok
- Faculty
of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu90220, Finland
| | - Harikanth Venkannagari
- Faculty
of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu90220, Finland
| | | | - Elena Dreassi
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, SienaI-53100, Italy
| | - Bernhard Lüscher
- Institute
of Biochemistry and Molecular Biology, RWTH
Aachen University, Aachen52074, Germany
| | - Patricia Korn
- Institute
of Biochemistry and Molecular Biology, RWTH
Aachen University, Aachen52074, Germany
| | - Oriana Tabarrini
- Department
of Pharmaceutical Sciences, University of
Perugia, Perugia06123, Italy,
| | - Lari Lehtiö
- Faculty
of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu90220, Finland,
| |
Collapse
|
5
|
Xu H, Mou YH, Guo MB, Zhang R, Yan ZZ, An R, Wang X, Su X, Hou Z, Guo C. Discovery of novel selenium-containing azole derivatives as antifungal agents by exploiting the hydrophobic cleft of CYP51. Eur J Med Chem 2022; 243:114707. [DOI: 10.1016/j.ejmech.2022.114707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 11/17/2022]
|
6
|
Guo MB, Yan ZZ, Wang X, Xu H, Guo C, Hou Z, Gong P. Design, synthesis and antifungal activities of novel triazole derivatives with selenium-containing hydrophobic side chains. Bioorg Med Chem Lett 2022; 78:129044. [DOI: 10.1016/j.bmcl.2022.129044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/17/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
|
7
|
Moghimi S, Shafiei M, Foroumadi A. Drug design strategies for the treatment azole-resistant candidiasis. Expert Opin Drug Discov 2022; 17:879-895. [PMID: 35793245 DOI: 10.1080/17460441.2022.2098949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Despite the availability of novel antifungals and therapeutic strategies, the rate of global mortality linked to invasive fungal diseases from fungal infection remains high. Candida albicans account for the most invasive mycosis produced by yeast. Thus, the current arsenal of medicinal chemists is focused on finding new effective agents with lower toxicity and broad-spectrum activity. In this review article, recent efforts to find effective agents against azole-resistant candidiasis, a common fungal infection, are covered. AREAS COVERED Herein, the authors outlined all azole-based compounds, dual target, and new scaffolds (non-azole-based compounds) which were effective against azole-resistant candidiasis. In addition, the mechanism of action and SAR studies were also discussed, if the data were available. EXPERT OPINION The current status of fungal infections and the drawbacks of existing drugs have encouraged scientists to find novel scaffolds based on different methods like virtual screening and fragment-based drug discovery. Machine learning and in-silico methods have found their role in this field and experts are hopeful to find novel scaffolds/compounds by using these methods.
Collapse
Affiliation(s)
- Setareh Moghimi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Shafiei
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Hou W, Dong H, Zhang X, Wang Y, Su L, Xu H. Selenium as an emerging versatile player in heterocycles and natural products modification. Drug Discov Today 2022; 27:2268-2277. [PMID: 35390546 DOI: 10.1016/j.drudis.2022.03.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/23/2022] [Accepted: 03/30/2022] [Indexed: 12/20/2022]
Abstract
The diverse pharmacological activities of organoselenium compounds are closely correlated to their ability to scavenge and induce reactive oxygen species (ROS), their intrinsic oxidative properties, and their Se(0) release property. The incorporation of selenium into small molecules, and particularly into heterocycles and natural products, has shown great potential in altering the potency and selectivity of these molecules. Therefore, selenium will play an important role in drug discovery in the near future. We summarize how different organoselenium species affect cellular oxidative stress levels, and try to correlate the structural properties of selenium-containing heterocycles and natural product derivatives to their biological activities and therapeutic applications. We also provide some information to guide the rational design of selenium-containing drugs.
Collapse
Affiliation(s)
- Wei Hou
- College of Pharmaceutical Science and Institute of Drug Development and Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Hewei Dong
- College of Pharmaceutical Science and Institute of Drug Development and Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xiang Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China.
| | - Yan Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Lin Su
- Hangzhou Minsheng Institutes for Pharma Research, Hangzhou 311121, China
| | - Hongtao Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
9
|
Hou W, Xu H. Incorporating Selenium into Heterocycles and Natural Products─From Chemical Properties to Pharmacological Activities. J Med Chem 2022; 65:4436-4456. [PMID: 35244394 DOI: 10.1021/acs.jmedchem.1c01859] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Selenium (Se)-containing compounds have emerged as potential therapeutic agents for the treatment of a range of diseases. Through tremendous effort, considerable knowledge has been acquired to understand the complex chemical properties and biological activities of selenium, especially after its incorporation into bioactive molecules. From this perspective, we compiled extensive literature evidence to summarize and critically discuss the relationship between the pharmacological activities and chemical properties of selenium compounds and the strategic incorporation of selenium into organic molecules, especially bioactive heterocycles and natural products. We also provide perspectives regarding the challenges in selenium-based medicinal chemistry and future research directions.
Collapse
Affiliation(s)
- Wei Hou
- College of Pharmaceutical Science and Institute of Drug Development and Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hongtao Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
10
|
Mistry S, Singh AK. Synthesis and in vitro antimicrobial activity of new steroidal hydrazone derivatives. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022. [DOI: 10.1186/s43094-021-00391-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
For many years, various drugs have been used for the treatment of infectious diseases but some bacterial microorganisms have induced resistance to several drugs. In a search of new antimicrobial agents, a series of new steroidal hydrazones were designed and synthesized.
Result
The structures of the compounds were established based on the spectral data. The in vitro antimicrobial activity of some newly synthesized compounds against bacteria and fungi was studied.
Conclusion
New compounds showed better or similar antimicrobial activity. Designing more efficient steroidal hydrazones from ketosteroid based on the current study may successfully lead to the development of antimicrobial agent.
Graphical abstract
Collapse
|
11
|
Xu H, Cao C, Wang X, Guo MB, Yan ZZ, An R, Zhang R, Dong EH, Mou YH, Hou Z, Guo C. Discovery of 1,2,3-selenadiazole analogues as antifungal agents using a scaffold hopping approach. Bioorg Chem 2021; 115:105182. [PMID: 34333426 DOI: 10.1016/j.bioorg.2021.105182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/25/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022]
Abstract
With the increasing incidence of antifungal resistance, new antifungal agents having novel scaffolds hence are in an urgent need to combat infectious diseases caused by multidrug-resistant (MDR) pathogens. In this study, we reported the design, synthesis, and pharmacological evaluation of novel 1,2,3-selenadiazole analogues by scaffold hopping strategy. Preliminary results of antifungal activity demonstrated that the new class of compounds showed broad-spectrum fungistatic and fungicidal activity. Most importantly, these newly synthesized compounds can eliminate these azole-resistant fungi and inhibit the formation of C. albicans biofilm. In particular, compound S07 showed promising antifungal activity against five azole-resistant strains with MIC values ranging from 4 to 32 μg/mL. Then, further target identification and mechanistic studies indicated that representative compound S07 exert its inhibitory activity by inhibiting fungal lanosterol 14α-demethylase enzyme (CYP51). Interestingly, representative compounds showed low cytotoxicity on mammalian cell lines. In addition, the molecular docking studies elucidated the binding modes of these compounds toward CYP51. Altogether, these results suggest that compound S07 with novel skeleton is a promising CYP51 inhibitor for treatment of fungal infections.
Collapse
Affiliation(s)
- Hang Xu
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Chun Cao
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Xin Wang
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Meng-Bi Guo
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Zhong-Zuo Yan
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Ran An
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Rui Zhang
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - En-Hui Dong
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Yan-Hua Mou
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhuang Hou
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China.
| | - Chun Guo
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China.
| |
Collapse
|
12
|
Musalov MV, Yakimov VA, Potapov VA, Zinchenko SV, Amosova SV. One-Pot Syntheses of Functionalized Dihydrobenzoselenophenes and Selenochromans from Acetyl Eugenol and Selenium Dibromide. Rearrangement of 2-(Bromomethyl)-2,3-dihydro-1-benzoselenophene to Selenochromans. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2021. [DOI: 10.1134/s1070428021040096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Xu H, Yan ZZ, Guo MB, An R, Wang X, Zhang R, Mou YH, Hou Z, Guo C. Lead optimization generates selenium-containing miconazole CYP51 inhibitors with improved pharmacological profile for the treatment of fungal infections. Eur J Med Chem 2021; 216:113337. [PMID: 33713977 DOI: 10.1016/j.ejmech.2021.113337] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 11/19/2022]
Abstract
A series of selenium-containing miconazole derivatives were identified as potent antifungal drugs in our previous study. Representative compound A03 (MIC = 0.01 μg/mL against C.alb. 5314) proved efficacious in inhibiting the growth of fungal pathogens. However, further study showed lead compound A03 exhibited potential hemolysis, significant cytotoxic effect and unfavorable metabolic stability and was therefore modified to overcome these drawbacks. In this article, the further optimization of selenium-containing miconazole derivatives resulted in the discovery of similarly potent compound B17 (MIC = 0.02 μg/mL against C.alb. 5314), exhibiting a superior pharmacological profile with decreased rate of metabolism, cytotoxic effect and hemolysis. Furthermore, compound B17 showed fungicidal activity against Candida albicans and significant effects on the treatment of resistant Candida albicans infections. Meanwhile, compound B17 not only could reduce the ergosterol biosynthesis pathway by inhibiting CYP51, but also inhibited biofilm formation. More importantly, compound B17 also shows promising in vivo efficacy after intraperitoneal injection and the PK study of compound B17 was evaluated. In addition, molecular docking studies provide a model for the interaction between the compound B17 and the CYP51 protein. Overall, we believe that these selenium-containing miconazole compounds can be further developed for the potential treatment of fungal infections.
Collapse
Affiliation(s)
- Hang Xu
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Zhong-Zuo Yan
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Meng-Bi Guo
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Ran An
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Xin Wang
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Rui Zhang
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China
| | - Yan-Hua Mou
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhuang Hou
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China.
| | - Chun Guo
- Key Laboratory of Structure-Based Drugs Design and Discovery (Ministry of Education), School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016 China.
| |
Collapse
|
14
|
Xu H, Su X, Guo MB, An R, Mou YH, Hou Z, Guo C. Design, synthesis, and biological evaluation of novel miconazole analogues containing selenium as potent antifungal agents. Eur J Med Chem 2020; 198:112360. [DOI: 10.1016/j.ejmech.2020.112360] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/17/2020] [Accepted: 04/17/2020] [Indexed: 12/19/2022]
|
15
|
Ma L, Wang H, Wang J, Liu L, Zhang S, Bu M. Novel Steroidal 5α,8α-Endoperoxide Derivatives with Semicarbazone/Thiosemicarbazone Side-chain as Apoptotic Inducers through an Intrinsic Apoptosis Pathway: Design, Synthesis and Biological Studies. Molecules 2020; 25:molecules25051209. [PMID: 32156024 PMCID: PMC7179397 DOI: 10.3390/molecules25051209] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
A series of novel steroidal 5α,8α-endoperoxide derivatives bearing semicarbazone (7a–g) or thiosemicarbazone (7h–k) side chain were designed, synthesized and evaluated for their cytotoxicities in four human cancer cell lines (HepG2, HCT-116, MCF-7, and A549) using the MTT assay in vitro. The results showed that compound 7j exhibited significant cytotoxic activity against HepG2 cells (IC50 = 3.52 μM), being more potent than ergosterol peroxide. Further cellular mechanism studies in HepG2 cells indicated that compound 7j triggered the mitochondrial-mediated apoptosis by decreasing mitochondrial membrane potential (MMP), which was associated with up-regulation of Bax, down-regulation of Bcl-2, activation levels of the caspase cascade, and formation of reactive oxygen species (ROS). The above findings indicated that compound 7j may be used as a promising skeleton for antitumor agents with improved efficacy.
Collapse
Affiliation(s)
- Liwei Ma
- Research Institute of Medicine & Pharmacy, Qiqihar Medical University, Qiqihar 161006, China;
| | - Haijun Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (H.W.); (J.W.); (L.L.)
| | - Jing Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (H.W.); (J.W.); (L.L.)
| | - Lei Liu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (H.W.); (J.W.); (L.L.)
| | - Song Zhang
- Basic Medical Science College, Qiqihar Medical University, Qiqihar 161006, China;
| | - Ming Bu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (H.W.); (J.W.); (L.L.)
- Correspondence: ; Tel.: +86-0452-266-3881
| |
Collapse
|
16
|
Zhang J, Sun X, Chen Y, Mi Y, Tan W, Miao Q, Li Q, Dong F, Guo Z. Preparation of 2,6-diurea-chitosan oligosaccharide derivatives for efficient antifungal and antioxidant activities. Carbohydr Polym 2020; 234:115903. [PMID: 32070523 DOI: 10.1016/j.carbpol.2020.115903] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/12/2020] [Accepted: 01/20/2020] [Indexed: 12/22/2022]
Abstract
In this study, 2-urea-chitosan oligosaccharide derivatives (2-urea-COS derivatives) and 2,6-diurea-chitosan oligosaccharide derivatives (2,6-diurea-COS derivatives) were successfully designed and synthesized via intermediate 2-methoxyformylated chitosan oligosaccharide. All samples were characterized and compared based on FT-IR, 1H NMR spectroscopy, and elemental analysis. The antifungal effects of COS derivatives were tested against Fusarium oxysporum f. sp. niveum, Phomopsis asparagus, and Botrytis cinereal. Their antioxidant properties, including superoxide radicals' scavenging activity, hydroxyl radicals' scavenging activity, and DPPH radicals' scavenging activity were also explored within different concentrations. COS derivatives bearing urea groups showed improved bioactivity compared with pristine COS and 2,6-diurea-COS derivatives had a higher biological activity than 2-urea-COS derivatives in tested concentrations. Additionally, L929 cells were used to carry out cytotoxicity test of COS and COS derivatives by CCK-8 assay. The results indicated that some of samples showed low cytotoxicity. These findings offered a suggestion that COS derivatives bearing urea groups are promising biological materials.
Collapse
Affiliation(s)
- Jingjing Zhang
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xueqi Sun
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuan Chen
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingqi Mi
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenqiang Tan
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China
| | - Qin Miao
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China
| | - Qing Li
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China
| | - Fang Dong
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China
| | - Zhanyong Guo
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|