1
|
Huo H, Dan W, Li M, Chen Y, Yang C, Wu L, Shi B, Li J. Design, synthesis, and biological evaluation of steroidal indole derivatives as membrane-targeting antibacterial candidates. Eur J Med Chem 2025; 283:117156. [PMID: 39671876 DOI: 10.1016/j.ejmech.2024.117156] [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: 09/24/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Rational modification of natural products plays a key role in drug discovery. Herein, a series of steroidal indole derivatives containing various substituents and steroidal skeletons were designed and synthesized with classical Fischer indole synthesis as a key step in an efficient synthetic route for the first time. The in vitro antibacterial activity of all the synthesized derivatives was evaluated against four Gram-positive strains including three Methicillin-Resistant Staphylococcus aureus. Compound 11e displayed the most potent antibacterial activity (MIC = 1-2 μg/mL) with low cytotoxicity and hemolytic activity. Derivative 11e displayed more rapid bactericidal kinetic than vancomycin in the time-kill study and was less likely to induce bacterial resistance. Moreover, the preliminary antibacterial mechanism explorations indicated that compound 11e could effectively inhibit biofilm formation, promote the accumulation of reactive oxygen species, decrease bacterial metabolism, and destroy bacterial cell membranes to exert its antibacterial effects. The study of in vivo antibacterial activity suggested that compound 11e could significantly reduce the bacteria counts in a mouse subcutaneous infection model. These findings provided a bright hope for steroidal indole derivatives as promising antibacterial candidates to settle drug resistance.
Collapse
Affiliation(s)
- Haibo Huo
- Department of Life Sciences, Changzhi University, Changzhi, 046011, Shanxi, China
| | - Wenjia Dan
- School of Life Science and Technology, Weifang Medical University, Shandong, China
| | - Min Li
- Department of Chemistry, Changzhi University, Changzhi, 046011, Shanxi, China
| | - Yanbin Chen
- Department of Life Sciences, Changzhi University, Changzhi, 046011, Shanxi, China
| | - Chaofu Yang
- School of Pharmacy, Changzhi Medical College, Changzhi, 046000, China
| | - Lintao Wu
- Department of Chemistry, Changzhi University, Changzhi, 046011, Shanxi, China.
| | - Baojun Shi
- College of Plant Protection, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Jian Li
- Department of Life Sciences, Changzhi University, Changzhi, 046011, Shanxi, China; Department of Chemistry, Changzhi University, Changzhi, 046011, Shanxi, China.
| |
Collapse
|
2
|
Liu JR, Jiang EY, Sukhbaatar O, Zhang WH, Zhang MZ, Yang GF, Gu YC. Natural and synthetic 5-(3'-indolyl)oxazoles: Biological activity, chemical synthesis and advanced molecules. Med Res Rev 2025; 45:97-143. [PMID: 39152525 DOI: 10.1002/med.22078] [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: 04/03/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/19/2024]
Abstract
5-(3'-Indolyl)oxazole moiety is a privileged heterocyclic scaffold, embedded in many biologically interesting natural products and potential therapeutic agents. Compounds containing this scaffold, whether from natural sources or synthesized, have demonstrated a wide array of biological activities. This has piqued the interest of synthetic chemists, leading to a large number of reported synthetic approaches to 5-(3'-indolyl)oxazole scaffold in recent years. In this review, we comprehensively overviewed the different biological activities and chemical synthetic methods for the 5-(3'-indolyl)oxazole scaffold reported in the literatures from 1963 to 2024. The focus of this study is to highlight the significance of 5-(3'-indolyl)oxazole derivatives as the lead compounds for the lead discovery of anticancer, pesticidal, antimicrobial, antiviral, antioxidant and anti-inflammatory agents, to summarize the synthetic methods for the 5-(3'-indolyl)oxazole scaffold. In addition, the reported mechanism of action of 5-(3'-indolyl)oxazoles and advanced molecules studied in animal models are also reviewed. Furthermore, this review offers perspectives on how 5-(3'-indolyl)oxazole scaffold as a privileged structure might be exploited in the future.
Collapse
Affiliation(s)
- Jing-Rui Liu
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing, China
| | - En-Yu Jiang
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing, China
| | - Otgonpurev Sukhbaatar
- Department of Chemistry, School of Applied Sciences, Mongolian University of Life Sciences, Ulaanbaatar, Mongolia
| | - Wei-Hua Zhang
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing, China
| | - Ming-Zhi Zhang
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing, China
| | - Guang-Fu Yang
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, China
| | - Yu-Cheng Gu
- Jealott's Hill International Research Centre, Syngenta, Bracknell, Berkshire, UK
| |
Collapse
|
3
|
Shamsi M, Al-Asbahy WM, Al-Areqi HQN, Alzowahi FAM. Probing the Biomolecular Interactions of DNA/HSA with the New Sn(IV) Complex and Computational Perspectives: Design, Synthesis, Characterization, Anticancer Activity, and Molecular Modeling Approach. J Med Chem 2024; 67:21841-21858. [PMID: 39661984 DOI: 10.1021/acs.jmedchem.4c01306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
The ligands 2,2'-bipyridyl and indole-3-carboxylic acid were used to create a Sn(IV) complex, which was then synthesized and carefully characterized using elemental analysis and spectroscopic techniques (UV-vis, IR, 1H, 13C, and 119Sn NMR, and ESI-MS) and RXPD. Utilizing biophysical techniques such as UV-vis, fluorescence titrations, circular dichroism, FTIR (for HSA), and cleavage activity (for DNA), in vitro binding studies of Sn(IV) complex and DNA/HSA were satisfied with the strong electrostatic binding interaction of the Sn(IV) complex via the phosphate backbone of the DNA helix as well as in the subdomain IIA of HSA. The observed trend in the binding interactions and computational studies of the Sn(IV) complex was attributed to the nature of the ligands bound to the Sn(IV) center that influences their in vitro activities. The Sn(IV) complex showed sufficient effectiveness to be considered a viable candidate for the creation of anticancer medications.
Collapse
Affiliation(s)
- Manal Shamsi
- Department of Pharmacy, Faculty of Medical Sciences, National University (Ibb Branch), Ibb 46654, Yemen
| | - Waddhaah M Al-Asbahy
- Department of Chemistry, Faculty of Applied Sciences, Taiz University, Taiz 6803, Yemen
| | - Hakim Q N Al-Areqi
- Department of Physics, Faculty of Applied Sciences, Taiz University, Taiz 6803, Yemen
| | - Fahad A M Alzowahi
- Department of Pharmacy, Faculty of Medical Sciences, National University (Ibb Branch), Ibb 46654, Yemen
| |
Collapse
|
4
|
Xu Y, Shao L, Zhou Z, Zhao L, Wan S, Sun W, Wanyan W, Yuan Y. ARG2 knockdown promotes G0/G1 cell cycle arrest and mitochondrial dysfunction in adenomyosis via regulation NF-κB and Wnt/Β-catenin signaling cascades. Int Immunopharmacol 2024; 140:112817. [PMID: 39116499 DOI: 10.1016/j.intimp.2024.112817] [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: 05/07/2024] [Revised: 06/28/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Adenomyosis is a common gynecological disease, characterized by overgrowth of endometrial glands and stroma in the myometrium, however its exact pathophysiology still remains uncertain. Emerging evidence has demonstrated the elevated level of arginase 2 (ARG2) in endometriosis and adenomyosis. This study aimed to determine whether ARG2 involved in mitochondrial function and epithelial to mesenchymal transition (EMT) in adenomyosis and its potential underlying mechanisms. MATERIALS AND METHODS RNA interference was used to inhibit ARG2 gene, and then Cell Counting Kit (CCK-8) assay and flow cytometery were performed to detect the cell proliferation capacity, cell cycle, and apoptosis progression, respectively. The mouse adenomyosis model was established and RT-PCR, Western blot analysis, mitochondrial membrane potential (Δψm) detection and mPTP opening evaluation were conducted. RESULTS Silencing ARG2 effectively down-regulated its expression at the mRNA and protein levels in endometrial cells, leading to decreased enzyme activity and inhibition of cell viability. Additionally, ARG2 knockdown induced G0/G1 cell cycle arrest, promoted apoptosis, and modulated the expression of cell cycle- and apoptosis-related regulators. Notably, the interference with ARG2 induces apoptosis by mitochondrial dysfunction, ROS production, ATP depletion, decreasing the Bcl-2/Bax ratio, releasing Cytochrome c, and increasing the expression of Caspase-9/-3 and PARP. In vivo study in a mouse model of adenomyosis demonstrated also elevated levels of ARG2 and EMT markers, while siARG2 treatment reversed EMT and modulated inflammatory cytokines. Furthermore, ARG2 knockdown was found to modulate the NF-κB and Wnt/β-catenin signaling pathways in mouse adenomyosis. CONCLUSION Consequently, ARG2 silencing could induce apoptosis through a mitochondria-dependent pathway mediated by ROS, and G0/G1 cell cycle arrest via suppressing NF-κB and Wnt/β-catenin signaling pathways in Ishikawa cells. These findings collectively suggest that ARG2 plays a crucial role in the pathogenesis of adenomyosis and may serve as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Yaping Xu
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China; State Key Laboratory of Ultrasound in Medicine and Engineering, No.1 Medical College Road, Yuzhong District, Chongqing, China
| | - Lin Shao
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China; State Key Laboratory of Ultrasound in Medicine and Engineering, No.1 Medical College Road, Yuzhong District, Chongqing, China
| | - Zhan Zhou
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Liying Zhao
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Shuquan Wan
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Wenjing Sun
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Wenya Wanyan
- Department of Gynaecology, Shandong Provincial Third Hospital, Shandong University, No.11 Wuyingshan Middle Road, Tianqiao District, Jinan, Shandong 250031, China
| | - Yinping Yuan
- State Key Laboratory of Ultrasound in Medicine and Engineering, No.1 Medical College Road, Yuzhong District, Chongqing, China; Department of Pathology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250031, China.
| |
Collapse
|
5
|
Zhang X, Liu G, Sun X, Wan LS, Zhou Y. A Metal-Free Direct Decarboxylative Fluoroacylation of Indole Carboxylic Acids with Fluorinated Acids. J Org Chem 2024; 89:14591-14595. [PMID: 39323110 DOI: 10.1021/acs.joc.4c01842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
A straightforward preparation of diversified fluorinated indol-3-yl ketones was developed by the direct decarboxylative fluoroacylation of indole carboxylic acids. The reaction could be performed on a gram scale under net conditions. Neither a metal catalyst nor an additive was employed. This methodology featured simple reaction conditions, high efficiency, exclusive selectivity, a broad substrate scope, and easy operation, which allowed it to meet the green chemistry requirement of the modern pharmaceutical industry. Control experiments confirmed that a radical process might be involved in the tandem decarboxylative fluoroacylation sequence.
Collapse
Affiliation(s)
- Xingxing Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guangyuan Liu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xing Sun
- Hebei Chemical and Pharmaceutical College, Shijiazhuang 050026, China
| | - Luo-Sheng Wan
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yirong Zhou
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
6
|
Rzepka Z, Bober-Majnusz K, Hermanowicz JM, Bębenek E, Chrobak E, Surażyński A, Wrześniok D. Assessment of the Lipophilicity of Indole Derivatives of Betulin and Their Toxicity in a Zebrafish Model. Molecules 2024; 29:4408. [PMID: 39339403 PMCID: PMC11434430 DOI: 10.3390/molecules29184408] [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: 08/06/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
There are scientific studies indicating that the attachment of an indole moiety to the triterpene scaffold can lead to increased anticancer potential. Lipophilicity is one of the factors that may influence biological properties and is therefore an important parameter to determine for newly obtained compounds as drug candidates. In the present study, previously synthesized 3 and/or 28-indole-betulin derivatives were evaluated for lipophilicity by reversed-phase thin-layer chromatography. The experimental values of lipophilicity (logPTLC) were then subjected to correlation analysis with theoretical values of logP, as well as for selected physicochemical and pharmacokinetic parameters and anticancer activity. A toxicity test using zebrafish embryos and larvae was also conducted. High correlation was observed between the experimental and theoretical values of lipophilicity. We presented correlation equations and statistical parameters describing the relationships between logPTLC and several physicochemical and ADME parameters. We also revealed the lack of correlation between the experimental values of lipophilicity and anticancer activity. Moreover, experiments on zebrafish have confirmed no toxicity of the tested compounds, which was consistent with the results of the in silico toxicity analysis. The results demonstrated, using the example of indole derivatives of betulin, the utility of lipophilicity values in the context of predicting the biological activity of new compounds.
Collapse
Affiliation(s)
- Zuzanna Rzepka
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska, 41-200 Sosnowiec, Poland;
| | - Katarzyna Bober-Majnusz
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland;
| | - Justyna Magdalena Hermanowicz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland;
- Department of Clinical Pharmacy, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
| | - Ewa Bębenek
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska, 41-200 Sosnowiec, Poland; (E.B.); (E.C.)
| | - Elwira Chrobak
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska, 41-200 Sosnowiec, Poland; (E.B.); (E.C.)
| | - Arkadiusz Surażyński
- Department of Medicinal Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland;
| | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska, 41-200 Sosnowiec, Poland;
| |
Collapse
|
7
|
Xu Y, Liang X, Hyun CG. Discovery of Indole-Thiourea Derivatives as Tyrosinase Inhibitors: Synthesis, Biological Evaluation, Kinetic Studies, and In Silico Analysis. Int J Mol Sci 2024; 25:9636. [PMID: 39273583 PMCID: PMC11394742 DOI: 10.3390/ijms25179636] [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: 07/29/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Tyrosinase, a key enzyme in melanin synthesis, represents a crucial therapeutic target for hyperpigmentation disorders due to excessive melanin production. This study aimed to design and evaluate a series of indole-thiourea derivatives by conjugating thiosemicarbazones with strong tyrosinase inhibitory activity to indole. Among these derivatives, compound 4b demonstrated tyrosinase inhibitory activity with an IC50 of 5.9 ± 2.47 μM, outperforming kojic acid (IC50 = 16.4 ± 3.53 μM). Kinetic studies using Lineweaver-Burk plots confirmed competitive inhibition by compound 4b. Its favorable ADMET and drug-likeness properties make compound 4b a promising therapeutic candidate with a reduced risk of toxicity. Molecular docking revealed that the compounds bind strongly to mushroom tyrosinase (mTYR) and human tyrosinase-related protein 1 (TYRP1), with compound 4b showing superior binding energies of -7.0 kcal/mol (mTYR) and -6.5 kcal/mol (TYRP1), surpassing both kojic acid and tropolone. Molecular dynamics simulations demonstrated the stability of the mTYR-4b complex with low RMSD and RMSF and consistent Rg and SASA values. Persistent strong hydrogen bonds with mTYR, along with favorable Gibbs free energy and MM/PBSA calculations (-19.37 kcal/mol), further support stable protein-ligand interactions. Overall, compound 4b demonstrated strong tyrosinase inhibition and favorable pharmacokinetics, highlighting its potential for treating pigmentary disorders.
Collapse
Affiliation(s)
- Yang Xu
- Jeju Inside Agency and Cosmetic Science Center, Department of Chemistry and Cosmetics, Jeju National University, Jeju 63243, Republic of Korea; (Y.X.); (X.L.)
| | - Xuhui Liang
- Jeju Inside Agency and Cosmetic Science Center, Department of Chemistry and Cosmetics, Jeju National University, Jeju 63243, Republic of Korea; (Y.X.); (X.L.)
| | - Chang-Gu Hyun
- Jeju Inside Agency and Cosmetic Science Center, Department of Chemistry and Cosmetics, Jeju National University, Jeju 63243, Republic of Korea; (Y.X.); (X.L.)
- Department of Beauty and Cosmetology, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
8
|
Ke J, Xie Y, Huang S, Wang W, Zhao Z, Lin W. Comparison of esophageal cancer survival after neoadjuvant chemoradiotherapy plus surgery versus definitive chemoradiotherapy: A systematic review and meta-analysis. Asian J Surg 2024; 47:3827-3840. [PMID: 38448293 DOI: 10.1016/j.asjsur.2024.02.099] [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: 08/10/2023] [Revised: 12/31/2023] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Surgery after neoadjuvant chemoradiotherapy remains the gold standard for the treatment of resectable esophageal cancer (EC); however, chemoradiotherapy without surgery has been recommended in specific cases. The aim of this meta-analysis is to analyse the survival between surgeries after neoadjuvant chemoradiotherapy compared with definitive chemoradiotherapy in order to provide a theoretical basis for clinically individualised differential treatment. We conducted an initial search of MEDLINE (PubMed), the Cochrane Library, and Embase for English-only articles that compared treatment regimens and provided survival data. According to the final I2 value of the two survival indicators, the random effect model or fixed effect model was used to calculate the overall hazard ratio (HR) and 95% confidence intervals (CI). Cochrane's Q test was used to judge the heterogeneity of the studies, and a funnel plot was used to evaluate for publication bias. A sensitivity analysis was performed to verify the stability of the included studies. A total of 38 studies involving 29161 patients (neoadjuvant therapy: 15401, definitive chemoradiotherapy: 13760) were included in the analysis. The final pooled results (HR = 0.74, 95% CI: 0.67-0.82) showed a statistically significant increase in overall survival with neoadjuvant chemoradiotherapy plus surgery compared with definitive chemoradiotherapy. Subgroup analyses were performed to determine the effects of heterogeneity, additional treatment regimens, study types, and geographic regions, as well as histologic differences, complications, and recurrence, on the overall results. For people with esophageal cancer that can be removed, neoadjuvant chemoradiotherapy combined with surgery improves survival compared to definitive chemoradiotherapy. However, more research is needed to confirm these results and help doctors make decisions about treatment.
Collapse
Affiliation(s)
- Junli Ke
- Department of Thoracic Surgery, Gaozhou People's Hospital Affiliated to Guangdong Medical University, Maoming, China
| | - Yujie Xie
- Department of Thoracic Surgery, Gaozhou People's Hospital Affiliated to Guangdong Medical University, Maoming, China
| | - Shenyang Huang
- Department of Cardiothoracic Surgery, Guangdong Medical University, Zhanjiang, China
| | - Wei Wang
- Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Zhengang Zhao
- Department of Cardiothoracic Surgery, Guangdong Medical University, Zhanjiang, China
| | - Wanli Lin
- Department of Thoracic Surgery, Gaozhou People's Hospital Affiliated to Guangdong Medical University, Maoming, China.
| |
Collapse
|
9
|
Zhang ZX, Zhang B, Yuan M, Zhao PF, Da CS. Pd(II)-Catalyzed C4-Selective Alkynylation of Indoles by a Transient Directing Group. Org Lett 2024; 26:6819-6824. [PMID: 39106047 DOI: 10.1021/acs.orglett.4c01970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
With alanine as a transient directing group, Pd-catalyzed regioselective alkynylation at the indole C4-position was successfully established in a good yield. The total synthesis of the PAF antagonist demonstrated the synthetic utility of this protocol. The regioselectivity was explicitly proven by the prepared C4-selective palladacycle intermediate in the catalytic process and the DFT calculation of the energy barriers of C4- and C2-site-selective C-H activation of indole.
Collapse
Affiliation(s)
- Ze-Xuan Zhang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Bing Zhang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Meng Yuan
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Peng-Fei Zhao
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Chao-Shan Da
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, P. R. China
- State Key Laboratory of Applied Organic Chemistry, Key Lab of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou 730000, P. R. China
| |
Collapse
|
10
|
Wang Z, Liu C, Huang J, Huang L, Feng H. Palladium-Catalyzed Regioselective Monofluoroallylation of Indoles with gem-Difluorocyclopropanes. Org Lett 2024; 26:6905-6909. [PMID: 39088798 DOI: 10.1021/acs.orglett.4c02554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
We present a palladium-catalyzed ring-opening reaction that induces indoles to cross-couple with gem-difluorocyclopropanes. The reaction proceeds through a domino process of C-C bond activation and C-F bond elimination, followed by C-C(sp2) coupling to produce various 2-fluoroallylindoles. This method is characterized by its high functional group tolerance, good yields and high regioselectivity, under base-free conditions. The synthetic utility of the products is illustrated by the functionalization of the NH and C2 positions of the indole scaffold.
Collapse
Affiliation(s)
- Zhenjie Wang
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Chuang Liu
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Junhai Huang
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Liliang Huang
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Huangdi Feng
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| |
Collapse
|
11
|
Yang Q, Li Q, Fan H. Antitumor activity of anlotinib in malignant melanoma: modulation of angiogenesis and vasculogenic mimicry. Arch Dermatol Res 2024; 316:447. [PMID: 38958761 DOI: 10.1007/s00403-024-03020-1] [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: 04/03/2024] [Revised: 04/03/2024] [Accepted: 04/26/2024] [Indexed: 07/04/2024]
Abstract
Malignant melanoma presents a formidable challenge due to its aggressive metastatic behavior and limited response to current treatments. To address this, our study delves into the impact of anlotinib on angiogenesis and vasculogenic mimicry using malignant melanoma cells and human umbilical vein endothelial cells. Evaluating tubular structure formation, cell proliferation, migration, invasion, and key signaling molecules in angiogenesis, we demonstrated that anlotinib exerts a dose-dependent inhibition on tubular structures and effectively suppresses cell growth and invasion in both cell types. Furthermore, in a mouse xenograft model, anlotinib treatment resulted in reduced tumor growth and vascular density. Notably, the downregulation of VEGFR-2, FGFR-1, PDGFR-β, and PI3K underscored the multitargeted antitumor activity of anlotinib. Our findings emphasize the therapeutic potential of anlotinib in targeting angiogenesis and vasculogenic mimicry, contributing to the development of novel strategies for combating malignant melanoma.
Collapse
MESH Headings
- Quinolines/pharmacology
- Quinolines/therapeutic use
- Quinolines/administration & dosage
- Humans
- Melanoma/drug therapy
- Melanoma/pathology
- Animals
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/pathology
- Indoles/pharmacology
- Indoles/therapeutic use
- Mice
- Xenograft Model Antitumor Assays
- Cell Proliferation/drug effects
- Human Umbilical Vein Endothelial Cells
- Cell Line, Tumor
- Vascular Endothelial Growth Factor Receptor-2/metabolism
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
- Cell Movement/drug effects
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Skin Neoplasms/drug therapy
- Skin Neoplasms/pathology
- Signal Transduction/drug effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/therapeutic use
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
- Mice, Nude
- Angiogenesis
Collapse
Affiliation(s)
- Qian Yang
- Department of Oncology and Hematology, People's Hospital of Leshan, 2-428 Yong'an Road, Leshan, 614000, Sichuan, People's Republic of China.
| | - Qianqian Li
- Department of General Medical, People's Hospital of Leshan, Leshan, 614000, People's Republic of China
| | - Hua Fan
- Department of Oncology and Hematology, People's Hospital of Leshan, 2-428 Yong'an Road, Leshan, 614000, Sichuan, People's Republic of China
| |
Collapse
|
12
|
Allawi MM, Razzak Mahmood AA, Tahtamouni LH, Saleh AM, Kanaan SI, Saleh KM, AlSakhen MF, Himsawi N, Yasin SR. Anti-proliferation evaluation of new derivatives of indole-6-carboxylate ester as receptor tyrosine kinase inhibitors. Future Med Chem 2024; 16:1313-1331. [PMID: 39109434 PMCID: PMC11318749 DOI: 10.1080/17568919.2024.2347084] [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: 01/27/2024] [Accepted: 04/10/2024] [Indexed: 08/10/2024] Open
Abstract
Aim: The main goal was to create two new groups of indole derivatives, hydrazine-1-carbothioamide (4a and 4b) and oxadiazole (5, and 6a-e) that target EGFR (4a, 4b, 5) or VEGFR-2 (6a-e). Materials & methods: The new derivatives were characterized using various spectroscopic techniques. Docking studies were used to investigate the binding patterns to EGFR/VEGFR-2, and the anti-proliferative properties were tested in vitro. Results: Compounds 4a (targeting EGFR) and 6c (targeting VEGFR-2) were the most effective cytotoxic agents, arresting cancer cells in the G2/M phase and inducing the extrinsic apoptosis pathway. Conclusion: The results of this study show that compounds 4a and 6c are promising cytotoxic compounds that inhibit the tyrosine kinase activity of EGFR and VEGFR-2, respectively.
Collapse
Affiliation(s)
- Mustafa M Allawi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Uruk university, Baghdad, Iraq
| | - Ammar A Razzak Mahmood
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Baghdad, Bab-Al-Mouadam, 10001, Baghdad, Iraq
| | - Lubna H Tahtamouni
- Department of Biology & Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
- Department of Biochemistry & Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Abdulrahman M Saleh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11884, Egypt
- Aweash El-Hagar Family Medicine Center, Epidemiological Surveillance Unit, MOHP, Mansoura, 35711, Egypt
| | - Sana I Kanaan
- Department of Biology & Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Khaled M Saleh
- Department of Biology & Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Mai F AlSakhen
- Department of Biology & Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Nisreen Himsawi
- Department of Microbiology, Pathology & Forensic Medicine, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Salem R Yasin
- Department of Biology & Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| |
Collapse
|
13
|
Barresi E, Baglini E, Poggetti V, Castagnoli J, Giorgini D, Salerno S, Taliani S, Da Settimo F. Indole-Based Compounds in the Development of Anti-Neurodegenerative Agents. Molecules 2024; 29:2127. [PMID: 38731618 PMCID: PMC11085553 DOI: 10.3390/molecules29092127] [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: 03/22/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Neurodegeneration is a gradual decay process leading to the depletion of neurons in both the central and peripheral nervous systems, ultimately resulting in cognitive dysfunctions and the deterioration of brain functions, alongside a decline in motor skills and behavioral capabilities. Neurodegenerative disorders (NDs) impose a substantial socio-economic strain on society, aggravated by the advancing age of the world population and the absence of effective remedies, predicting a negative future. In this context, the urgency of discovering viable therapies is critical and, despite significant efforts by medicinal chemists in developing potential drug candidates and exploring various small molecules as therapeutics, regrettably, a truly effective treatment is yet to be found. Nitrogen heterocyclic compounds, and particularly those containing the indole nucleus, which has emerged as privileged scaffold, have attracted particular attention for a variety of pharmacological applications. This review analyzes the rational design strategy adopted by different research groups for the development of anti-neurodegenerative indole-based compounds which have the potential to modulate various molecular targets involved in NDs, with reference to the most recent advances between 2018 and 2023.
Collapse
Affiliation(s)
- Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.B.); (V.P.); (J.C.); (F.D.S.)
| | - Emma Baglini
- Institute of Clinical Physiology, National Research Council of Italy, CNR Research Area, 56124 Pisa, Italy;
| | - Valeria Poggetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.B.); (V.P.); (J.C.); (F.D.S.)
| | - Jacopo Castagnoli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.B.); (V.P.); (J.C.); (F.D.S.)
| | - Doralice Giorgini
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, Fisciano, 84084 Salerno, Italy;
| | - Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.B.); (V.P.); (J.C.); (F.D.S.)
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.B.); (V.P.); (J.C.); (F.D.S.)
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (E.B.); (V.P.); (J.C.); (F.D.S.)
| |
Collapse
|
14
|
Yao CH, Wu MH, Chang PW, Wu SH, Song JS, Huang HH, Chen YC, Lee JC. Design, synthesis, and anticancer evaluation of 1-benzo[1,3]dioxol-5-yl-3-N-fused heteroaryl indoles. Mol Divers 2024; 28:595-608. [PMID: 36735167 DOI: 10.1007/s11030-023-10605-x] [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: 11/28/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023]
Abstract
A series of 1-benzo[1,3]dioxol-5-yl-indoles bearing 3-N-fused heteroaryl moieties have been designed based on literature reports of the activity of indoles against various cancer cell lines, synthesized via a Pd-catalyzed C-N cross-coupling, and evaluated for their anticancer activity against prostate (LNCaP), pancreatic (MIA PaCa-2), and acute lymphoblastic leukemia (CCRF-CEM) cancer cell lines. A detailed structure-activity relationship study culminated in the identification of 3-N-benzo[1,2,5]oxadiazole 17 and 3-N-2-methylquinoline 20, whose IC50 values ranged from 328 to 644 nM against CCRF-CEM and MIA PaCa-2. Further mechanistic studies revealed that 20 caused cell cycle arrest at the S phase and induced apoptosis in CCRF-CEM cancer cells. These 1-benzo[1,3]dioxol-5-yl-3-N-fused heteroaryl indoles may serve as a template for further optimization to afford more active analogs and develop a comprehensive understanding of the structure-activity relationships of indole anticancer molecules.
Collapse
Affiliation(s)
- Chun-Hsu Yao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Mine-Hsine Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Po-Wei Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Szu-Huei Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Jen-Shin Song
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Hsing-Hao Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Chun Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Jinq-Chyi Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan.
| |
Collapse
|
15
|
Wang Q, Wen W, Zhou L, Liu F, Ren X, Yu L, Chen H, Jiang Z. LL-37 improves sepsis-induced acute lung injury by suppressing pyroptosis in alveolar epithelial cells. Int Immunopharmacol 2024; 129:111580. [PMID: 38310763 DOI: 10.1016/j.intimp.2024.111580] [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: 11/03/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND LL-37 (also known as murine CRAMP) is a human antimicrobial peptide that plays a crucial role in innate immune defence against sepsis through various mechanisms. However, its involvement in sepsis-induced lung injury remains unclear. OBJECTIVES This work investigates the impact of LL-37 on pyroptosis generated by LPS in alveolar epithelial cells. The research utilizes both in vivo and in vitro sepsis-associated acute lung injury (ALI) models to understand the underlying molecular pathways. METHODS In vivo, an acute lung injury model induced by sepsis was established by intratracheal administration of LPS in C57BL/6J mice, which were subsequently treated with low-dose CRAMP (recombinant murine cathelicidin, 2.5 mg.kg-1) and high-dose CRAMP (5.0 mg.kg-1). In vitro, pyroptosis was induced in a human alveolar epithelial cell line (A549) by stimulation with LPS and ATP. Treatment was carried out with recombinant human LL-37, or LL-37 was knocked out in A549 cells using small interfering RNA (siRNA). Subsequently, haematoxylin and eosin staining was performed to observe the histopathological changes in lung tissues in the control group and sepsis-induced lung injury group. TUNEL and PI staining were used to observe DNA fragmentation and pyroptosis in mouse lung tissues and cells in the different groups. An lactate dehydrogenase (LDH) assay was performed to measure the cell death rate. The expression levels of NLRP3, caspase1, caspase 1 p20, GSDMD, NT-GSDMD, and CRAMP were detected in mice and cells using Western blotting, qPCR, and immunohistochemistry. ELISA was used to assess the levels of interleukin (IL)-1β and IL-18 in mouse serum, bronchoalveolar lavage fluid (BALF) and lung tissue and cell culture supernatants. RESULTS The expression of NLRP3, caspase1 p20, NT-GSDMD, IL 18 and IL1β in the lung tissue of mice with septic lung injury was increased, which indicated activation of the canonical pyroptosis pathway and coincided with an increase in CRAMP expression. Treatment with recombinant CRAMP improved pyroptosis in mice with lung injury. In vitro, treatment with LPS and ATP upregulated these classic pyroptosis molecules, LL-37 knockdown exacerbated pyroptosis, and recombinant human LL-37 treatment alleviated pyroptosis in alveolar epithelial cells. CONCLUSION These findings indicate that LL-37 protects against septic lung injury by modulating the expression of classic pyroptotic pathway components, including NLRP3, caspase1, and GSDMD and downstream inflammatory factors in alveolar epithelial cells.
Collapse
Affiliation(s)
- Quanzhen Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Wei Wen
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Lei Zhou
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, China; Department of Respiratory Intensive Care Unit, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Fen Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Xiaoxu Ren
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Lifeng Yu
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China
| | - Huanqin Chen
- Department of Gerontology, Qilu Hospital, Shandong University, Jinan, 250012 Shandong, China
| | - Zhiming Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong 250014, China.
| |
Collapse
|
16
|
Chuang YT, Yen CY, Shiau JP, Chang FR, Duh CY, Sung PJ, Chen KL, Tsai YH, Tang JY, Jeng JH, Sheu JH, Chang HW. Demethoxymurrapanine, an indole-naphthoquinone alkaloid, inhibits the proliferation of oral cancer cells without major side effects on normal cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:1221-1234. [PMID: 37921086 DOI: 10.1002/tox.24002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/23/2023] [Accepted: 10/07/2023] [Indexed: 11/04/2023]
Abstract
Antioral cancer drugs need a greater antiproliferative impact on cancer than on normal cells. Demethoxymurrapanine (DEMU) inhibits proliferation in several cancer cells, but an in-depth investigation was necessary. This study evaluated the proliferation-modulating effects of DEMU, focusing on oral cancer and normal cells. DEMU (0, 2, 3, and 4 μg/mL) at 48 h treatments inhibited the proliferation of oral cancer cells (the cell viability (%) for Ca9-22 cells was 100.0 ± 2.2, 75.4 ± 5.6, 26.0 ± 3.8, and 15.4 ± 1.4, and for CAL 27 cells was 100.0 ± 9.4, 77.2 ± 5.9, 57.4 ± 10.7, and 27.1 ± 1.1) more strongly than that of normal cells (the cell viability (%) for S-G cells was 100.0 ± 6.6, 91.0 ± 4.6, 95.0 ± 2.6, and 95.8 ± 5.5), although this was blocked by the antioxidant N-acetylcysteine. The presence of oxidative stress was evidenced by the increase of reactive oxygen species and mitochondrial superoxide and the downregulation of the cellular antioxidant glutathione in oral cancer cells, but these changes were minor in normal cells. DEMU also caused greater induction of the subG1 phase, extrinsic and intrinsic apoptosis (annexin V and caspases 3, 8, and 9), and DNA damage (γH2AX and 8-hydroxy-2-deoxyguanosine) in oral cancer than in normal cells. N-acetylcysteine attenuated all these DEMU-induced changes. Together, these data demonstrate the preferential antiproliferative function of DEMU in oral cancer cells, with the preferential induction of oxidative stress, apoptosis, and DNA damage in these cancer cells, and low cytotoxicity toward normal cells.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei, Taiwan
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan, Taiwan
| | - Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chang-Yih Duh
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ping-Jyun Sung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- National Museum of Marine Biology and Aquarium, Pingtung, Taiwan
| | - Kuan-Liang Chen
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yi-Hong Tsai
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jiiang-Huei Jeng
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Jyh-Horng Sheu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
17
|
Aputen AD, Elias MG, Gilbert J, Sakoff JA, Gordon CP, Scott KF, Aldrich-Wright JR. Platinum(IV) Prodrugs Incorporating an Indole-Based Derivative, 5-Benzyloxyindole-3-Acetic Acid in the Axial Position Exhibit Prominent Anticancer Activity. Int J Mol Sci 2024; 25:2181. [PMID: 38396859 PMCID: PMC10888562 DOI: 10.3390/ijms25042181] [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: 11/29/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Kinetically inert platinum(IV) complexes are a chemical strategy to overcome the impediments of standard platinum(II) antineoplastic drugs like cisplatin, oxaliplatin and carboplatin. In this study, we reported the syntheses and structural characterisation of three platinum(IV) complexes that incorporate 5-benzyloxyindole-3-acetic acid, a bioactive ligand that integrates an indole pharmacophore. The purity and chemical structures of the resultant complexes, P-5B3A, 5-5B3A and 56-5B3A were confirmed via spectroscopic means. The complexes were evaluated for anticancer activity against multiple human cell lines. All complexes proved to be considerably more active than cisplatin, oxaliplatin and carboplatin in most cell lines tested. Remarkably, 56-5B3A demonstrated the greatest anticancer activity, displaying GI50 values between 1.2 and 150 nM. Enhanced production of reactive oxygen species paired with the decline in mitochondrial activity as well as inhibition of histone deacetylase were also demonstrated by the complexes in HT29 colon cells.
Collapse
Affiliation(s)
- Angelico D. Aputen
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (A.D.A.); (M.G.E.); (C.P.G.)
| | - Maria George Elias
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (A.D.A.); (M.G.E.); (C.P.G.)
- Ingham Institute, Sydney, NSW 2170, Australia;
| | - Jayne Gilbert
- Calvary Mater Newcastle Hospital, Newcastle, NSW 2298, Australia; (J.G.); (J.A.S.)
| | - Jennette A. Sakoff
- Calvary Mater Newcastle Hospital, Newcastle, NSW 2298, Australia; (J.G.); (J.A.S.)
| | - Christopher P. Gordon
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (A.D.A.); (M.G.E.); (C.P.G.)
| | - Kieran F. Scott
- Ingham Institute, Sydney, NSW 2170, Australia;
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Janice R. Aldrich-Wright
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (A.D.A.); (M.G.E.); (C.P.G.)
| |
Collapse
|
18
|
Li F, Wang X, Cai Y, Lin Y, Tang Y, Wang S. Gut microbiota-derived metabolites as novel therapies for inflammatory bowel diseases: Role of nuclear receptors. FUNDAMENTAL RESEARCH 2024. [DOI: 10.1016/j.fmre.2024.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
19
|
Chen Y, Lu Z, He W, Zhu H, Lu W, Shi J, Sheng J, Xie W. Rhodium-catalyzed annulation of hydrazines with vinylene carbonate to synthesize unsubstituted 1-aminoindole derivatives. RSC Adv 2024; 14:4804-4809. [PMID: 38323018 PMCID: PMC10844929 DOI: 10.1039/d3ra07466h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/30/2024] [Indexed: 02/08/2024] Open
Abstract
Herein, we describe rhodium-catalysed C-H bond activation for [3 + 2] annulation using hydrazide and vinylene carbonate, providing an efficient method for synthesising unsubstituted 1-aminoindole compounds. Characterised by high yields, mild reaction conditions, and no need for external oxidants, this transformation demonstrates excellent regioselectivity and a wide tolerance for various functional groups.
Collapse
Affiliation(s)
- Yichun Chen
- School of Environment and Chemical Engineering, Foshan University Foshan 528000 China
| | - Ziqi Lu
- School of Environment and Chemical Engineering, Foshan University Foshan 528000 China
| | - Wenfen He
- School of Environment and Chemical Engineering, Foshan University Foshan 528000 China
| | - Huanyi Zhu
- School of Environment and Chemical Engineering, Foshan University Foshan 528000 China
| | - Weilong Lu
- School of Environment and Chemical Engineering, Foshan University Foshan 528000 China
| | - Junjun Shi
- School of Environment and Chemical Engineering, Foshan University Foshan 528000 China
| | - Jie Sheng
- School of Environment and Chemical Engineering, Foshan University Foshan 528000 China
| | - Wucheng Xie
- School of Environment and Chemical Engineering, Foshan University Foshan 528000 China
| |
Collapse
|
20
|
Tian K, Deng B, Han X, Zheng H, Lin T, Wang Z, Zhang Y, Wang G. Over-expression of microRNA-145 Elevating Autophagy Activities via Downregulating FRS2 Expression. Comb Chem High Throughput Screen 2024; 27:127-135. [PMID: 37264620 DOI: 10.2174/1386207326666230602090848] [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: 10/04/2022] [Revised: 04/11/2023] [Accepted: 04/25/2023] [Indexed: 06/03/2023]
Abstract
OBJECTIVES Osteoarthritis (OA) is one of the most common chronic and progressive joint diseases characterized by cartilage degeneration and chondrocyte death. In this study, we aimed to identify the modulation effect of miR-145 on chondrocytes' autophagy during the development of OA. BACKGROUND Osteoarthritis (OA) is one of the most prevalent types of chronic and progressive joint disorder with the symptoms of joint pain and stiffness, and it leads to disability at the end stage. In recent years, microRNA-145 (miR-145) has been found to activate autophagy in various cell types, including mesenchymal stem cells, cardiomyocytes, and osteosarcoma cells. However, it is unknown whether miR-145 regulates the progression of OA by influencing chondrocyte autophagy. METHODS Before investigating the regulatory effect of miR-145 on the autophagic activity of chondrocytes, the expression of miR-145 in human joint samples was analyzed. The targeting relationship between miR-145 and FRS2 was detected by dual luciferase assay. The effect of FRS2 and miR-145 on the autophagic activity of chondrocytes was observed by bidirectional expression of FRS2 and miR-145. RESULTS The miR-145 expression and LC3-II/LC3-I ratio were significantly decreased and the SQSTM1 expression was increased in OA patients. The miR-145 overexpression in C20A4 cells increased LC3-II/LC3-I ratio, decreased SQSTM1 expression, and was positively correlated with autophagic activity. Under oxidative stress, miR-145 overexpression significantly improved chondrocyte viability through autophagy stimulation. FRS2 is a potential target of miR-145 via a binding sequence within its 3' UTR. FRS2 acts as the downstream mediator of miR-145 to suppress autophagy through activating PI3K/Akt/mTOR pathways. CONCLUSION The miR-145 acts as a protective factor against chondrocytes by regulating miRFRS2- autophagy axis. The decrease of miR-145 in articular synovial fluid may turn out to be an important marker for early diagnosis of OA, and modulation of miR-145 may represent a promising therapeutic strategy for OA.
Collapse
Affiliation(s)
- Ke Tian
- Department of Orthopedics and Joint, Affiiated Hospital of Jining Medical University, Shandong, 272001, China
| | - Bin Deng
- Department of Orthopedics, Affiliated Hospital of Jining Medical University, Shandong Province, Zoucheng District, Jining, 273500, Shandong, People's Republic of China
| | - Xiaodong Han
- Department of Orthopedics, Affiliated Hospital of Jining Medical University, Shandong Province, Zoucheng District, Jining, 273500, Shandong, People's Republic of China
| | - Haiyi Zheng
- Department of Orthopedics and Joint, Affiiated Hospital of Jining Medical University, Shandong, 272001, China
| | - Tao Lin
- Department of Orthopedics and Joint, Affiiated Hospital of Jining Medical University, Shandong, 272001, China
| | - Zhimeng Wang
- Department of Orthopedics and Joint, Affiiated Hospital of Jining Medical University, Shandong, 272001, China
| | - Yuanmin Zhang
- Department of Orthopedics and Joint, Affiiated Hospital of Jining Medical University, Shandong, 272001, China
| | - Guodong Wang
- Department of Orthopedics and Joint, Affiiated Hospital of Jining Medical University, Shandong, 272001, China
| |
Collapse
|
21
|
Fu Q, Zhang F, Vijayalakshmi A. The Protective Effect of Sanggenol L Against DMBA-induced Hamster Buccal Pouch Carcinogenesis Induces Apoptosis and Inhibits Cell Proliferative Signalling Pathway. Comb Chem High Throughput Screen 2024; 27:885-893. [PMID: 37496247 DOI: 10.2174/1386207326666230726140706] [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: 12/17/2022] [Revised: 05/21/2023] [Accepted: 06/09/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) has a poor prognosis when treated with surgery and chemotherapy. Therefore, a new therapy and preventative strategy for OSCC and its underlying mechanisms are desperately needed. The purpose of this study was to examine the chemopreventive effects of sanggenol L on oral squamous cell carcinoma (OSCC). The research focused on molecular signalling pathways in 7,12-dimethylbenz(a)anthracene (DMBA)-induced hamster buccal pouch (HBP) carcinogenesis. AIM The purpose of this study was to look at the biochemical and chemopreventive effects of sanggenol L on 7,12-dimethylbenz(a)anthracene (DMBA)-induced HBP (hamster buccal pouch) carcinogenesis via cell proliferation and the apoptotic pathway. METHODS After developing squamous cell carcinoma, oral tumours continued to progress leftward into the pouch 3 times per week for 10 weeks while being exposed to 0.5 % reactive DMBA three times per week. Tumour growth was caused by biochemical abnormalities that induced inflammation, increased cell proliferation, and decreased apoptosis. RESULTS Oral sanggenol L (10 mg/kg bw) supplementation with cancer-induced model DMBApainted hamsters prevented tumour occurrences, improved biochemistry, inhibited inflammatory markers, decreased cell proliferation marker expression of tumour necrosis factor-alpha (TNF- α), nuclear factor (NF-κB), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and induced apoptosis. CONCLUSION Sanggenol L could be developed into a new medicine for the treatment of oral carcinogenesis.
Collapse
Affiliation(s)
- Qing Fu
- Department of Stomatology, People's Hospital of Qijiang District, Chongqing, 401420, China
| | - Fangming Zhang
- Department of Stomatology, The Fifth People's Hospital Of Wuxi, Wuxi, 214000, China
| | - Annamalai Vijayalakshmi
- Department of Biochemistry, Rabiammal Ahamed Maideen College for Women, Thiruvarur, Tamil Nadu, 610001, India
| |
Collapse
|
22
|
Wen S, Hu M, Chen C, Li Z, Liu G. Neuritin Alleviates Diabetic Retinopathy by Regulating Endoplasmic Reticulum Stress in Rats. Comb Chem High Throughput Screen 2024; 27:2454-2461. [PMID: 38173210 DOI: 10.2174/0113862073275316231123060640] [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: 09/07/2023] [Revised: 11/06/2023] [Accepted: 11/16/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Neuritin, a small-molecule neurotrophic factor, maintains neuronal cell activity, inhibits apoptosis, promotes process growth, and regulates neural progenitor cell differentiation, migration, and synaptic maturation. Neuritin helps retinal ganglion cells (RGCs) survive optic nerve injury in rats and regenerate axons. However, the role of Neuritin in Diabetic retinopathy (DR) is unclear. OBJECTIVE This study is intended to investigate the effect and mechanism of Neuritin in DR. For this purpose, we established DR rat models and injected Neuritin into them. This study provides a potential treatment for diabetic retinopathy. METHODS The rat model of DR was established by streptozotocin (STZ) injection, and the effect of Neuritin on DR was detected by intravitreal injection. Histological analysis was performed by H&E and TUNEL methods. The mRNA and protein expressions of endoplasmic reticulum stress (ERS) pathway-related transcription factors were detected by qRT-PCR and western blot. The blood-retinal barrier (BRB) function was assessed using the patch-clamp technique and Evans blue leakage assay. RESULTS Neuritin significantly improved the retinal structure, restrained the apoptosis of retinal cells, and protected the normal function of BRB in DR model rats. Mechanistically, Neuritin may function by inhibiting the expression of GRP78, ASK1, Caspase-12, VEGF, and so on. CONCLUSION Our results indicate that Neuritin alleviates retinal damage in DR rats via the inactive endoplasmic reticulum pathway. Our study provides a potential treatment for DR.
Collapse
Affiliation(s)
- Shu Wen
- Department of Ophthalmology, Jingmen No. 1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China
| | - Meng Hu
- Department of Ophthalmology, Jingmen No. 1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China
| | - Changzheng Chen
- Renmin Hospital of Wuhan University, Hubei General Hospital, Wuhan, Hubei, 430000, China
| | - Zhen Li
- Department of Ophthalmology, Jingmen No. 1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China
| | - Guoli Liu
- Department of Ophthalmology, Jingmen No. 1 People's Hospital, No. 168, Xiangshan Avenue, Duodao District, Jingmen, 448000, China
| |
Collapse
|
23
|
Li J, Jiang Q, Wang X, Hou L, Wang L, Lou K, Pang S. Metformin Preserves Insulin Secretion in Pancreatic β-cells through FGF21/Akt Pathway In vitro and In vivo. Comb Chem High Throughput Screen 2024; 27:2691-2698. [PMID: 37855357 DOI: 10.2174/0113862073246747230920170201] [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: 03/03/2023] [Revised: 06/09/2023] [Accepted: 08/06/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND In our previous studies, it was found that metformin can elevate the expression of FGF21 in the peripheral blood of type 2 diabetic rats and improve insulin sensitivity in diabetic rats. However, whether this effect is mediated by increased FGF21 expression in pancreatic islet β-cells is still unknown. Therefore, this study focuses on the effect of metformin on insulin secretion in pancreatic β-cells. AIMS Metformin can effectivly improve insulin resistance. Metformin influencing pancreatic β- cell function is inclusive. In this study, we sought to analyze possible variations in insulin secretion and possible signaling mechanisms after metformin intervention. METHODS The study employed an in vivo model of a high-fat diet in streptozocin-induced diabetic rats and an in vitro model of rat pancreatic β-cells (INS-1 cells) that were subjected to damage caused by hyperglycemia and hyperlipidemia. After treating INS-1 cells in normal, high-glucose, and high-glucose+metformin, we measured insulin secretion by glucose-stimulated insulin secretion (GSIS). Insulin was measured using an enzyme-linked immunosorbent assay. FGF21 expression was detected by RT-PCR and Western blot, as well as that p-Akt and t-Akt expression were detected by Western blot in INS-1 cells and diabetic rat islets. Finally, to verify the regulation of the FGF21 /Akt axis in metformin administration, additional experiments were carried out in metformin-stimulated INS-1 cells. RESULTS High-glucose could significantly stimulate insulin secretion while metformin preserved insulin secretion. Expression of FGF21 and p-Akt was decreased in high-glucose, however, metformin could reverse this effect in INS-1 cells and diabetic rat islets. CONCLUSION Our results demonstrate a protective role of metformin in preserving insulin secretion through FGF21/Akt signaling in T2DM.
Collapse
Affiliation(s)
- Jianting Li
- Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Qiang Jiang
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Xin Wang
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Lulu Hou
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Lulu Wang
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Kai Lou
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
| | - Shuguang Pang
- Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, 250012, China
- Department of Endocrinology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, 250021, China
- Department of Clinical Medicine, Weifang Medical College, Weifang, 261000, China
| |
Collapse
|
24
|
Rohilla S, Goyal G, Berwal P, Mathur N. A Review on Indole-triazole Molecular Hybrids as a Leading Edge in Drug Discovery: Current Landscape and Future Perspectives. Curr Top Med Chem 2024; 24:1557-1588. [PMID: 38766822 DOI: 10.2174/0115680266307132240509065351] [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: 02/24/2024] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 05/22/2024]
Abstract
Molecular hybridization is a rational design strategy used to create new ligands or prototypes by identifying and combining specific pharmacophoric subunits from the molecular structures of two or more known bioactive derivatives. Molecular hybridization is a valuable technique in drug discovery, enabling the modulation of unwanted side effects and the creation of potential dual-acting drugs that combine the effects of multiple therapeutic agents. Indole-triazole conjugates have emerged as promising candidates for new drug development. The indole and triazole moieties can be linked through various synthetic strategies, such as click chemistry or other coupling reactions, to generate a library of diverse compounds for biological screening. The achievable structural diversity with indole-triazole conjugates offers avenues to optimize their pharmacokinetic and pharmacodynamic attributes, amplifying their therapeutic efficacy. Researchers have extensively tailored both indole and triazole frameworks with diverse modifications to comprehend their impact on the drug's pharmacokinetic and pharmacodynamic characteristics. The current review article endeavours to explore and discuss various research strategies to design indoletriazole hybrids and elucidate their significance in a variety of pathological conditions. The insights provided herein are anticipated to be beneficial for the researchers and will likely encourage further exploration in this field.
Collapse
Affiliation(s)
- Suman Rohilla
- Department of Pharmaceutical Chemistry, SGT College of Pharmacy, Shree Guru Gobind Singh Tricentenary University, Gurugram, Haryana, India
| | - Garima Goyal
- Department of Pharmaceutical Chemistry, SGT College of Pharmacy, Shree Guru Gobind Singh Tricentenary University, Gurugram, Haryana, India
| | - Paras Berwal
- Department of Pharmaceutical Chemistry, SGT College of Pharmacy, Shree Guru Gobind Singh Tricentenary University, Gurugram, Haryana, India
| | - Nancy Mathur
- Department of Pharmaceutical Chemistry, SGT College of Pharmacy, Shree Guru Gobind Singh Tricentenary University, Gurugram, Haryana, India
| |
Collapse
|
25
|
Lin H, Ma C, Zhong A, Zang H, Chen W, Li L, Le Y, Xie Q. Anti-Angiogenic Agents Combined with Immunotherapy for Advanced Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Comb Chem High Throughput Screen 2024; 27:1081-1091. [PMID: 37559541 DOI: 10.2174/1386207326666230808112656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/17/2023] [Accepted: 07/03/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND Anti-angiogenic agents could enhance tumor immunity response, and anti- angiogenesis plus immunotherapy has become a novel treatment option for advanced non-small cell lung cancer (NSCLC). The efficacy of this combination therapy remains controversial and obscure. AIM We conducted a meta-analysis to evaluate the clinical efficacy and safety of this therapeutic strategy in patients with advanced NSCLC and provide more guidance for treating NSCLC clinically. METHODS A systematic literature search was performed in PubMed, Embase, Web of Science, CNKI, and Wanfang databases to identify relevant studies published up to December 2021. The primary endpoint was the objective response rate (ORR). Second endpoints were progression-free survival (PFS), overall survival (OS), and grade ≥3 AEs adverse events (AEs). The sensitivity analysis was conducted to confirm the stability of the results. STATA 15.0 was utilized for all pooled analyses. RESULTS Eleven studies were eventually included in the meta-analysis, involving 533 patients with advanced NSCLC. The pooled ORR rate was 27% (95% CI 18% to 35%; I2 =84.2%; p<0.001), while the pooled median PFS and OS was 5.84 months (95% CI 4.66 to 7.03 months; I2=78.4%; p<0.001) and 14.20 months (95% CI 11.08 to 17.32 months; I2=82.2%; p=0.001), respectively. Most common grade ≥3 AEs included hypertension, hand-foot syndrome, diarrhea, adrenal insufficiency, hyponatremia, proteinuria, rash, thrombocytopenia, and fatigue. CONCLUSION Anti-angiogenesis combined with immunotherapy demonstrated satisfactory antitumor activity and an acceptable toxicity profile in patients with advanced NSCLC. The pooled results of our meta-analysis provided further evidence supporting the favorable efficacy and safety of this therapeutic strategy.
Collapse
Affiliation(s)
- Heng Lin
- Department of Oncology, Fuzhou Pulmonary Hospital of Fujian, Fuzhou, Fujian, 350008, China
| | - Chenhui Ma
- Department of Thoracis Surgery, Fuzhou Pulmonary Hospital of Fujian, Fuzhou, Fujian, 350008, China
| | - Aihong Zhong
- Department of Oncology, Fuzhou Pulmonary Hospital of Fujian, Fuzhou, Fujian, 350008, China
| | - Huanping Zang
- Department of Oncology, Fuzhou Pulmonary Hospital of Fujian, Fuzhou, Fujian, 350008, China
| | - Wenxin Chen
- Department of Oncology, Fuzhou Pulmonary Hospital of Fujian, Fuzhou, Fujian, 350008, China
| | - Lixiu Li
- Department of Oncology, Fuzhou Pulmonary Hospital of Fujian, Fuzhou, Fujian, 350008, China
| | - Yuyin Le
- Department of Oncology, Fuzhou Pulmonary Hospital of Fujian, Fuzhou, Fujian, 350008, China
| | - Qiang Xie
- Department of Oncology, Fuzhou Pulmonary Hospital of Fujian, Fuzhou, Fujian, 350008, China
| |
Collapse
|
26
|
Li X, Chen S, Zheng G, Yang Y, Yin N, Niu X, Yao L, Lv P. Atorvastatin Calcium Ameliorates Cognitive Deficits Through the AMPK/Mtor Pathway in Rats with Vascular Dementia. Comb Chem High Throughput Screen 2024; 27:148-156. [PMID: 37282650 DOI: 10.2174/1386207326666230606114448] [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: 02/21/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/08/2023]
Abstract
AIM In this study, the protective effects of atorvastatin calcium (AC) on nerve cells and cognitive improvement in vivo and in vitro were investigated by establishing cell models and vascular dementia (VD) rat models. BACKGROUND VD is a neurodegenerative disease characterized by cognitive deficits caused by chronic cerebral hypoperfusion. AC has been studied for its potential to cure VD but its efficacy and underlying mechanism are still unclear. OBJECTIVE The mechanism of action of AC on cognitive deficits in the early stages of VD is unclear. Here, the 2-vessel occlusion (2-VO) model in vivo and the hypoxia/reoxygenation (H/R) cell model in vitro was established to investigate the function of AC in VD. METHODS The spatial learning and memory abilities of rats were detected by the Morris method. The IL-6, tumour necrosis factor-α (TNF-α), malondialdehyde (MDA) and superoxide dismutase (SOD) in cell supernatant was tested by ELISA kits. After behavioural experiments, rats were anaesthetized and sacrificed, and their brains were extracted. One part was immediately fixed in 4% paraformaldehyde for H&E, Nissl, and immunohistochemical analyses, and the other was stored in liquid nitrogen. All data were shown as mean ± SD. Statistical comparison between the two groups was performed by Student's t-test. A two-way ANOVA test using GraphPad Prism 7 was applied for escape latency analysis and the swimming speed test. The difference was considered statistically significant at p < 0.05. RESULTS AC decreased apoptosis, increased autophagy, and alleviated oxidative stress in primary hippocampal neurons. AC regulated autophagy-related proteins in vitro by western blotting. VD mice improved cognitively in the Morris water maze. Spatial probing tests showed that VD animals administered AC had considerably longer swimming times to the platform than VD rats. H&E and Nissl staining showed that AC reduces neuronal damage in VD rats. Western blot and qRT-PCR indicated that AC in VD rats inhibited Bax and promoted LC3-II, Beclin-1, and Bcl-2 in the hippocampus region. AC also improves cognition via the AMPK/mTOR pathway. CONCLUSION This study found that AC may relieve learning and memory deficits as well as neuronal damage in VD rats by changing the expression of apoptosis/autophagy-related genes and activating the AMPK/mTOR signalling pathway in neurons.
Collapse
Affiliation(s)
- Xiuqin Li
- Department of Neurology, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
- Department of Geriatrics, Hebei General Hospital, Shijiazhuang, Hebei, 050051, China
| | - Shaopeng Chen
- Department of Preventive Health, Hebei General Hospital, Shijiazhuang, Hebei, 050051, China
| | - Guiming Zheng
- Department of Rheumatology and Immunology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, China
| | - Yanyan Yang
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, China
| | - Nan Yin
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, China
| | - Xiaoli Niu
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, China
| | - Lixia Yao
- Department of Geriatrics, Hebei General Hospital, Shijiazhuang, Hebei, 050051, China
| | - Peiyuan Lv
- Department of Neurology, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, 050051, China
| |
Collapse
|
27
|
Thirunavukkarasu MK, Veerappapillai S, Karuppasamy R. Sequential virtual screening collaborated with machine-learning strategies for the discovery of precise medicine against non-small cell lung cancer. J Biomol Struct Dyn 2024; 42:615-628. [PMID: 36995235 DOI: 10.1080/07391102.2023.2194994] [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: 12/16/2022] [Accepted: 03/17/2023] [Indexed: 03/31/2023]
Abstract
Dysregulation of MAPK pathway receptors are crucial in causing uncontrolled cell proliferation in many cancer types including non-small cell lung cancer. Due to the complications in targeting the upstream components, MEK is an appealing target to diminish this pathway activity. Hence, we have aimed to discover potent MEK inhibitors by integrating virtual screening and machine learning-based strategies. Preliminary screening was conducted on 11,808 compounds using the cavity-based pharmacophore model AADDRRR. Further, seven ML models were accessed to predict the MEK active compounds using six molecular representations. The LGB model with morgan2 fingerprints surpasses other models ensuing 0.92 accuracy and 0.83 MCC value versus test set and 0.85 accuracy and 0.70 MCC value with external set. Further, the binding ability of screened hits were examined using glide XP docking and prime-MM/GBSA calculations. Note that we have utilized three ML-based scoring functions to predict the various biological properties of the compounds. The two hit compounds such as DB06920 and DB08010 resulted excellent binding mechanism with acceptable toxicity properties against MEK. Further, 200 ns of MD simulation combined with MM-GBSA/PBSA calculations confirms that DB06920 may have stable binding conformations with MEK thus step forwarded to the experimental studies in the near future.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Muthu Kumar Thirunavukkarasu
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Shanthi Veerappapillai
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Ramanathan Karuppasamy
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
28
|
Shi L, Li H, Sun L, Tian C, Li H. Alleviation of Angiotensin II-Induced Vascular Endothelial Cell Injury Through Long Non-coding RNA TUG1 Inhibition. Comb Chem High Throughput Screen 2024; 27:1523-1532. [PMID: 37818575 DOI: 10.2174/0113862073265220231004071645] [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: 06/27/2023] [Revised: 07/28/2023] [Accepted: 08/18/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Hypertension damages endothelial cells, causing vascular remodelling. It is caused by Ang II-induced endothelial cell (EC) destruction. The long noncoding RNA (lncRNAs) are emerging regulators of endothelium homeostasis. Injured endothelium expresses lncRNA taurine-upregulated gene 1 (TUG1), which may mediate endothelial cell damage, proliferation, apoptosis, and autophagy and contribute to cardiovascular disease. However, uncertainty surrounds the function of lncRNA TUG1, on arterial endothelium cell damage. OBJECTIVE This research aimed to investigate the role and mechanism of lncRNA TUG1 in vascular endothelial cell injury. METHOD A microarray analysis of lncRNA human gene expression was used to identify differentially expressed lncRNAs in human umbilical vein endothelial cell (HUVEC) cultures. The viability, apoptosis, and migration of Ang II-treated HUVECs were then evaluated. In order to investigate the role of lncRNA TUG1 in hypertension, qRT-PCR, western blotting, and RNA-FISH were used to examine the expression of TUG1 in SHR mice. RESULTS Ang II-activated HUVECs and SHR rats' abdominal aortas highly express the lncRNA TUG1. LncRNA TUG1 knockdown in HUVECs could increase cell viability, reduce apoptosis, and produce inflammatory factors. In SHR rat abdominal aortas, lncRNA TUG1 knockdown promoted proliferation and inhibited apoptosis. HE spotting showed that lncRNA TUG1 knockdown improved SHR rats' abdominal aorta shape. lncRNA TUG1 knockdown promotes miR-9- 5p, which inhibits CXCR4 following transcription. The lncRNA TUG1/miR-9-5p/CXCR4 axis and vascular cell injury were also examined. MiR-9-5p silencing or CXCR4 overexpression lowered cell survival, apoptosis, and lncRNA TUG1-induced IL-6 and NO expression. CONCLUSION lncRNA TUG1 suppression could reduce Ang II-induced endothelial cell damage by regulating and targeting miR-9-5p to limit CXCR4 expression and open new vascular disease research pathways.
Collapse
Affiliation(s)
- Lin Shi
- Department of Internal Medicine-Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 16369 Jing Shi Road, Li Xi District, Jinan, Shandong, 250014, China
| | - Hui Li
- Department of Emergency Internal Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 16369 Jing Shi Road, Li Xi District, Jinan, Shandong, 250014, China
| | - Lingzhi Sun
- Department of Internal Medicine-Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 16369 Jing Shi Road, Li Xi District, Jinan, Shandong, 250014, China
| | - Caijun Tian
- Department of Internal Medicine-Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 16369 Jing Shi Road, Li Xi District, Jinan, Shandong, 250014, China
| | - Haitao Li
- Department of Internal Medicine-Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 16369 Jing Shi Road, Li Xi District, Jinan, Shandong, 250014, China
| |
Collapse
|
29
|
Mishra S, Sahu A, Kaur A, Kaur M, Kumar J, Wal P. Recent Development in the Search for Epidermal Growth Factor Receptor (EGFR) Inhibitors based on the Indole Pharmacophore. Curr Top Med Chem 2024; 24:581-613. [PMID: 37909440 DOI: 10.2174/0115680266264206231020111820] [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: 06/21/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 11/03/2023]
Abstract
The signal transduction and cell proliferation are regulated by the epidermal growth factor receptor. The proliferation of tumor cells, apoptosis, invasion, and angiogenesis is inhibited by the epidermal growth factor receptor. Thus, breast cancer, non-small cell lung cancer, cervical cancer, glioma, and bladder cancer can be treated by targeting the epidermal growth factor receptor. Although third-generation epidermal growth factor receptor inhibitors are potent drugs, patients exhibit drug resistance after treatment. Thus, the search for new drugs is being continued. Among the different potent epidermal growth factor receptor inhibitors, we have reviewed the indole-based inhibitors. We have discussed the structure-activity relationship of the compounds with the active sites of the epidermal growth factor receptor receptors, their synthesis, and molecular docking studies.
Collapse
Affiliation(s)
- Shweta Mishra
- SGT College of Pharmacy, SGT University, Gurugram, Haryana, 122505, India
| | - Adarsh Sahu
- Department of Pharmaceutical Sciences, Dr. Hari Singh Gour Vishwavidyalaya (A Central University), Sagar, 473003, Madhya Pradesh, India
- Amity Institute of Pharmacy, Amity University Rajasthan, NH11C Kant Kanwar Jaipur, 300202, India
| | - Avneet Kaur
- SGT College of Pharmacy, SGT University, Gurugram, Haryana, 122505, India
| | | | - Jayendra Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology, Delhi-NCR Campus, Ghaziabad, UP, 201204, India
| | - Pranay Wal
- Pranveer Singh Institute of Technology, Pharmacy, Kanpur, UP, India
| |
Collapse
|
30
|
Dalal N, Makharia GK, Dalal M, Mohan A, Singh R, Kumar A. Gut Metabolite Indoxyl Sulfate Has Selective Deleterious and Anticancer Effect on Colon Cancer Cells. J Med Chem 2023; 66:17074-17085. [PMID: 38103027 DOI: 10.1021/acs.jmedchem.3c01907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
There are a number of reports about anticancer activity of indole derivatives. In this study, we investigated the role of indoxyl sulfate (IS) for its selective anticancer activity on colon cancer cells. IS treatment on HCT-116 and HT-29 human epithelial adenocarcinoma cells led to a decrease in cell proliferation, cell viability, and ATP content. Colon cancer cells showed a 10% increase in cell apoptosis in comparison to control. Due to IS treatment, cell morphology got distorted, cell number found decreased, intracellular vesicles formed, and cells were found floating in the media. Cells also showed a loss in membrane integrity and a decrease in colony-forming ability and ceased at the G2/M phase of the cell cycle. No significant change was noted in the level of inflammatory cytokines IL-17A, IL-1β, and TNF-α, histology, length of intestine, and spleen after 100 mM IS treatment to balb/c mice. These observations indicate the selective anticancer effect of IS on colon cancer cells.
Collapse
Affiliation(s)
- Nishu Dalal
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India
- Department of Environmental Studies, Satyawati College, Delhi University, Delhi 110052, India
| | - Govind K Makharia
- Department of Gastroenterology and Human Nutrition, AIIMS, New Delhi 110029, India
| | - Monu Dalal
- ICMR - National Institute of Malaria Research, New Delhi 110077, India
| | - Anand Mohan
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Rajeev Singh
- Department of Environmental Studies, Satyawati College, Delhi University, Delhi 110052, India
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India
| |
Collapse
|
31
|
Duan SF, Song L, Guo HY, Deng H, Huang X, Shen QK, Quan ZS, Yin XM. Research status of indole-modified natural products. RSC Med Chem 2023; 14:2535-2563. [PMID: 38107170 PMCID: PMC10718587 DOI: 10.1039/d3md00560g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 12/19/2023] Open
Abstract
Indole is a heterocyclic compound formed by the fusion of a benzene ring and pyrrole ring, which has rich biological activity. Many indole-containing compounds have been sold on the market due to their excellent pharmacological activity. For example, vincristine and reserpine have been widely used in clinical practice. The diverse structures and biological activities of natural products provide abundant resources for the development of new drugs. Therefore, this review classifies natural products by structure, and summarizes the research progress of indole-containing natural product derivatives, their biological activities, structure-activity relationship and research mechanism which has been studied in the past 13 years, so as to provide a basis for the development of new drug development.
Collapse
Affiliation(s)
- Song-Fang Duan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Lei Song
- Yanbian University Hospital, Yanbian University Yanji 133002 People's Republic of China
| | - Hong-Yan Guo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Hao Deng
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Xing Huang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Qing-Kun Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Zhe-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Xiu-Mei Yin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| |
Collapse
|
32
|
Singh G, Priyanka, Sushma, Sharma S, Deep Kaur J, Devi A, Gupta S, Devi S, Mohan B. Designing of efficient two-armed colorimetric and fluorescent indole appended organosilicon sensors for the detection of Al(III) ions: Implication as paper-based sensor. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 302:123015. [PMID: 37364410 DOI: 10.1016/j.saa.2023.123015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/28/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023]
Abstract
Metal ions have significant roles in diagnosis, industry, human health, and the environment. To design and develop new lucid molecular receptors for the selective detection of metal ions is important for environmental and medical applications. In the present work, two-armed indole appended Schiff bases conjoined with 1,2,3-Triazole bis-organosilane and bis-organosilatrane skelton sensors for naked eye colorimetric and fluorescent detection sensors for Al(III) are developed. The introduction of Al(III) in sensor 4 and 5 show red shift in UV-visible spectra, changes in fluorescence spectra and immediate color change from colorless to dark yellow. Furthermore, the pH and time response studies were explored for both sensors 4 & 5. The sensors 4 and 5 exhibited significantly low detection limit (LOD) in nano-molar range 1.41 × 10-9 M and 0.17 × 10-9 M respectively from emission titration. The LOD form absorption titration was found to be 0.6 × 10-7 M for sensor 4 and 0.22 × 10-7 M for sensor 5. In addition, the sensing model is developed as paper based sensor for its practical applicability. The theoretical calculations were performed on Gaussian 03 program by relaxing the structures using Density functional theory.
Collapse
Affiliation(s)
- Gurjaspreet Singh
- Department of Chemistry and Centre of Advanced Studies Panjab University, Chandigarh 160014, India.
| | - Priyanka
- Department of Chemistry and Centre of Advanced Studies Panjab University, Chandigarh 160014, India.
| | - Sushma
- Department of Chemistry and Centre of Advanced Studies Panjab University, Chandigarh 160014, India
| | - Sanjay Sharma
- Department of Chemistry and Centre of Advanced Studies Panjab University, Chandigarh 160014, India
| | - Jashan Deep Kaur
- Department of Chemistry and Centre of Advanced Studies Panjab University, Chandigarh 160014, India
| | - Anita Devi
- Department of Chemistry and Centre of Advanced Studies Panjab University, Chandigarh 160014, India
| | - Sofia Gupta
- Department of Chemistry and Centre of Advanced Studies Panjab University, Chandigarh 160014, India
| | - Swati Devi
- Department of Chemistry and Centre of Advanced Studies Panjab University, Chandigarh 160014, India
| | - Brij Mohan
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
33
|
Salem MG, Abu El-Ata SA, Elsayed EH, Mali SN, Alshwyeh HA, Almaimani G, Almaimani RA, Almasmoum HA, Altwaijry N, Al-Olayan E, Saied EM, Youssef MF. Novel 2-substituted-quinoxaline analogs with potential antiproliferative activity against breast cancer: insights into cell cycle arrest, topoisomerase II, and EGFR activity. RSC Adv 2023; 13:33080-33095. [PMID: 37954422 PMCID: PMC10633821 DOI: 10.1039/d3ra06189b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023] Open
Abstract
Breast cancer is a global health concern, with increasing disease burden and disparities in access to healthcare. Late diagnosis and limited treatment options in underserved areas contribute to poor outcomes. In response to this challenge, we developed a novel family of 2-substituted-quinoxaline analogues, combining coumarin and quinoxaline scaffolds known for their anticancer properties. Through a versatile synthetic approach, we designed, synthesized, and characterized a set of 2-substituted quinoxaline derivatives. The antiproliferative activity of the synthesized compounds was assessed toward the MCF-7 breast cancer cells. Our investigations showed that the synthesized compounds exhibit considerable antiproliferative activity toward MCF-7 cells. Notably, compound 3b, among examined compounds, displayed a superior inhibitory effect (IC50 = 1.85 ± 0.11 μM) toward the growth of MCF-7 cells compared to the conventional anticancer drug staurosporine (IC50 = 6.77 ± 0.41 μM) and showed minimal impact on normal cells (MCF-10A cell lines, IC50 = 33.7 ± 2.04 μM). Mechanistic studies revealed that compound 3b induced cell cycle arrest at the G1 transition and triggered apoptosis in MCF-7 cells, as evidenced by increasing the percentage of cells arrested in the G2/M and pre-G1 phases utilizing flow cytometric analysis and Annexin V-FITC/PI analysis. Moreover, compound 3b was found to substantially suppress topoisomerase enzyme activity in MCF-7 cells. Molecular modeling studies further supported the potential of compound 3b as a therapeutic candidate by demonstrating significant binding affinity to the active sites of both topoisomerase II and EGFR proteins. Taken together, the presented 2-substituted-quinoxaline analogues, especially compound 3b, show promise as potential candidates for the development of effective anti-breast cancer drugs.
Collapse
Affiliation(s)
- Manar G Salem
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Suez Canal University Ismailia 41522 Egypt
| | - Sara A Abu El-Ata
- Department of Chemistry, Faculty of Science, Port Said University Port Said Egypt
| | - Elsherbiny H Elsayed
- Department of Chemistry, Faculty of Science, Port Said University Port Said Egypt
| | - Suraj N Mali
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology Ranchi 835215 India
| | - Hussah Abdullah Alshwyeh
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University Dammam 31441 Saudi Arabia
- Basic & Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University PO Box 1982 Dammam 31441 Saudi Arabia
| | - Ghassan Almaimani
- Department of Surgery, Faculty of Medicine, Umm Al-Qura University Al Abdeyah, PO Box 7607 Makkah Saudi Arabia
| | - Riyad A Almaimani
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University Al Abdeyah, PO Box 7607 Makkah Saudi Arabia
| | - Hussain A Almasmoum
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University Al Abdeyah, PO Box 7607 Makkah Saudi Arabia
| | - Najla Altwaijry
- Department of Pharmaceutical Sciences, Princess Nourah Bint Abdulrahman University PO Box 84428 Riyadh 11671 Saudi Arabia
| | - Ebtesam Al-Olayan
- Department of Zoology, College of Science, King Saud University Riyadh Saudi Arabia
| | - Essa M Saied
- Department of Chemistry (Biochemistry Division), Faculty of Science, Suez Canal University Ismailia 41522 Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin Brook-Taylor-Str. 2 12489 Berlin Germany
| | - Mohamed F Youssef
- Department of Chemistry (Organic Chemistry Division), Faculty of Science, Suez Canal University Ismailia 41522 Egypt
| |
Collapse
|
34
|
Liu M, Xie D, Hu D, Zhang R, Wang Y, Tang L, Zhou B, Zhao B, Yang L. In Situ Cocktail Nanovaccine for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207697. [PMID: 37740439 PMCID: PMC10625102 DOI: 10.1002/advs.202207697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 06/22/2023] [Indexed: 09/24/2023]
Abstract
In situ vaccination is a desirable strategy for cancer immunotherapy due to its convenience and capacity to target tumor antigens. Here, an in situ nanovaccine based on a cationic peptide with cholesterol-modified, DP7-C, for cancer immunotherapy is rationally designed, and developed a cancer nanovaccine that is easy to preparate. The nanovaccine includes cocktail small interfering RNAs (siRNAs) and immunologic adjuvant CpG ODNs, has synergistic effect in the cancer treatment. This nanovaccine can induce tumor cell death, promote antigen presentation and relieve immune suppression in the tumor microenvironment (TME). Moreover, this nanovaccine is administered to CT26 (hot) and B16F10 (cold) tumor model mice, in which it targeted the primary tumors and induced systemic antitumor immunity to inhibit metastasis. It is validated that the nanovaccine can convert cold tumors into hot tumors. Furthermore, the nanovaccine increased the immune response to anti-PD-1 therapy by modulating the TME in both CT26- and B16F10-tumor-bearing mice. The siRNA cocktail/CpG ODN/self-assembling peptide nanovaccine is a simple and universal tool that can effectively generate specific tumor cell antigens and can be combined with immuno-oncology agents to enhance antitumor immune activity. The versatile methodology provides an alternative approach for developing cancer nanovaccines.
Collapse
Affiliation(s)
- Mohan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Daoyuan Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Die Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bailing Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Binyan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
35
|
Fayad E, Altalhi SA, Abualnaja MM, Alrohaimi AH, Elsaid FG, Abu Almaaty AH, Saleem RM, Bazuhair MA, Ahmed Maghrabi AH, Beshay BY, Zaki I. Novel Acrylate-Based Derivatives: Design, Synthesis, Antiproliferative Screening, and Docking Study as Potential Combretastatin Analogues. ACS OMEGA 2023; 8:38394-38405. [PMID: 37867686 PMCID: PMC10586439 DOI: 10.1021/acsomega.3c05051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023]
Abstract
A variety of 3-(4-chlorophenyl) acrylic acids 4a,b and 3-(4-chlorophenyl)acrylate esters 5a-i were synthesized and structurally proven by spectroscopic studies such as IR, 1H NMR, and 13C NMR as well as mass spectrometry. All substances were investigated for their antiproliferative efficacy against the MDA-MB-231 cell line. Among these, acrylic acid compound 4b demonstrated the most potent cytotoxic effect with an IC50 value of 3.24 ± 0.13 μM, as compared to CA-4 (IC50 = 1.27 ± 09 μM). Additionally, acrylic acid molecule 4b displayed an inhibitory effect against β-tubulin polymerization with a percentage inhibition of 80.07%. Furthermore, compound 4b was found to produce considerable cell cycle arrest at the G2/M stage and cellular death, as demonstrated by FACS analysis. In addition, the in vivo antitumor screening of the sodium salt of acrylic acid 4b was carried out, and the results have shown that the tested molecule showed a significant decrease in viable EAC count and EAC volume, accompanied by a considerable increase in the life span prolongation, if compared to the positive control group. Furthermore, molecular modeling studies were performed to understand how the highly efficient chemicals 4b and 5e interact with the colchicine-binding region on tubulin. This work aims to shed light on the reasons behind their exceptional cytotoxicity and their better capacity to inhibit tubulin in comparison to CA-4.
Collapse
Affiliation(s)
- Eman Fayad
- Department
of Biotechnology, College of Sciences, Taif
University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Sarah Awwadh Altalhi
- Department
of Biotechnology, College of Sciences, Taif
University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Matokah M. Abualnaja
- Department
of Chemistry, Faculty of Applied Science, Umm Al-Qura University, Makkah
Al Mukarrama 24230, Saudi Arabia
| | - Abdulmohsen H. Alrohaimi
- Department
of Pharmacy Practice, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Fahmy G. Elsaid
- Biology
Department, College of Science, King Khalid
University, P.O.Box 960, Asir, Abha 61421, Saudi Arabia
| | - Ali H. Abu Almaaty
- Zoology
Department, Faculty of Science Port Said
University, Port Said 42526, Egypt
| | - Rasha Mohammed Saleem
- Department
of Laboratory Medicine, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha 65431, Saudi Arabia
| | - Mohammed A. Bazuhair
- Department
of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ali Hassan Ahmed Maghrabi
- Department
of Biology, Faculty of Applied Science, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Botros Y. Beshay
- Pharmaceutical
Sciences (Pharmaceutical Chemistry) Department, College of Pharmacy, Arab Academy for Science, Technology and Maritime
Transport, Alexandria 21913, Egypt
| | - Islam Zaki
- Pharmaceutical
Organic Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt
| |
Collapse
|
36
|
Goel B, Jaiswal S, Jain SK. Indole derivatives targeting colchicine binding site as potential anticancer agents. Arch Pharm (Weinheim) 2023; 356:e2300210. [PMID: 37480173 DOI: 10.1002/ardp.202300210] [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: 04/13/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/23/2023]
Abstract
Microtubules are appealing as intracellular targets for anticancer activity due to their importance in cell division. Three important binding sites are present on the tubulin protein: taxane, vinca, and colchicine binding sites (CBS). Many USFDA-approved drugs such as paclitaxel, ixabepilone, vinblastine, and combretastatin act by altering the dynamics of the microtubules. Additionally, a large number of compounds have been synthesized by medicinal chemists around the globe that target different tubulin binding sites. Although CBS inhibitors have proved their cytotoxic potential, no CBS-targeting drug had been able to reach the market. Several studies have reported design, synthesis, and biological evaluation of indole derivatives as potential anticancer agents. These compounds have been shown to inhibit cancer cell proliferation, induce apoptosis, and disrupt microtubule formation. Moreover, the binding affinity of these compounds to the CBS has been demonstrated using molecular docking studies and competitive binding assays. The present work has reviewed indole derivatives as potential colchicine-binding site inhibitors. The structure-activity relationship studies have revealed the crucial pharmacophoric features required for the potent and selective binding of indole derivatives to the CBS. The development of these compounds with improved efficacy and reduced toxicity could potentially lead to the development of novel and effective cancer therapies.
Collapse
Affiliation(s)
- Bharat Goel
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Shivani Jaiswal
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Shreyans K Jain
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| |
Collapse
|
37
|
Li Y, Liu Y, Zhang D, Chen J, Yang G, Tang P, Yang C, Liu J, Zhang J, Ouyang L. Discovery, Synthesis, and Evaluation of Novel Dual Inhibitors of a Vascular Endothelial Growth Factor Receptor and Poly(ADP-Ribose) Polymerase for BRCA Wild-Type Breast Cancer Therapy. J Med Chem 2023; 66:12069-12100. [PMID: 37616488 DOI: 10.1021/acs.jmedchem.3c00640] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors have been approved for the treatment of breast cancer (BC) with breast cancer susceptibility (BRCA) gene mutation. Leveraging new synthetic lethal interactions may be an effective way to broaden the indication of PARP inhibitors for BC patients with wild-type BRCA. Vascular endothelial growth factor receptor (VEGFR)-mediated suppression of angiogenesis has been reported to improve the sensitivity of wild-type BRCA cells to PARP inhibitors through synthetic lethality. Herein, we reported the conjugation of a PARP inhibitor with a VEGFR inhibitor pharmacophore to construct dual VEGFR and PARP inhibitors. The most potent compound 14b is identified to exert promising activities against VEGFR and PARP in the nanomolar range and possesses significant in vitro and in vivo antitumor and antimetastasis features. It also presented a favorable pharmacokinetic characteristics in rats with an oral bioavailability of 60.1%. Collectively, 14b may be a promising therapeutic agent of BRCA wild-type BC.
Collapse
Affiliation(s)
- Yang Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yun Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dan Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Juncheng Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gaoxia Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Pan Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengcan Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jifa Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
| | - Liang Ouyang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy and Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China
| |
Collapse
|
38
|
Xie X, Laster KV, Li J, Nie W, Yi YW, Liu K, Seong YS, Dong Z, Kim DJ. OSGIN1 is a novel TUBB3 regulator that promotes tumor progression and gefitinib resistance in non-small cell lung cancer. Cell Mol Life Sci 2023; 80:272. [PMID: 37646890 PMCID: PMC11071769 DOI: 10.1007/s00018-023-04931-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/26/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Oxidative stress induced growth inhibitor 1 (OSGIN1) regulates cell death. The role and underlying molecular mechanism of OSGIN1 in non-small cell lung cancer (NSCLC) are uncharacterized. METHODS OSGIN1 expression in NSCLC samples was detected using immunohistochemistry and Western blotting. Growth of NSCLC cells and gefitinib-resistant cells expressing OSGIN1 or TUBB3 knockdown was determined by MTT, soft agar, and foci formation assays. The effect of OSGIN1 knockdown on in vivo tumor growth was assessed using NSCLC patient-derived xenograft models and gefitinib-resistant patient-derived xenograft models. Potentially interacting protein partners of OSGIN1 were identified using IP-MS/MS, immunoprecipitation, PLA, and Western blotting assays. Microtubule dynamics were explored by tubulin polymerization assay and immunofluorescence. Differential expression of signaling molecules in OSGIN1 knockdown cells was investigated using phospho-proteomics, KEGG analysis, and Western blotting. RESULTS We found that OSGIN1 is highly expressed in NSCLC tissues and is positively correlated with low survival rates and tumor size in lung cancer patients. OSGIN1 knockdown inhibited NSCLC cell growth and patient-derived NSCLC tumor growth in vivo. Knockdown of OSGIN1 strongly increased tubulin polymerization and re-established gefitinib sensitivity in vitro and in vivo. Additionally, knockdown of TUBB3 strongly inhibited NSCLC cell proliferation. Mechanistically, we found that OSGIN1 enhances DYRK1A-mediated TUBB3 phosphorylation, which is critical for inducing tubulin depolymerization. The results of phospho-proteomics and ontology analysis indicated that knockdown of OSGIN1 led to reduced propagation of the MKK3/6-p38 signaling axis. CONCLUSIONS We propose that OSGIN1 modulates microtubule dynamics by enhancing DYRK1A-mediated phosphorylation of TUBB3 at serine 172. Moreover, elevated OSGIN1 expression promotes NSCLC tumor growth and gefitinib resistance through the MKK3/6-p38 signaling pathway. Our findings unveil a new mechanism of OSGIN1 and provide a promising therapeutic target for NSCLC treatment in the clinic.
Collapse
Affiliation(s)
- Xiaomeng Xie
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, 450008, Henan, China
- China-US (Henan) Hormel Cancer Institute, No, 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Kyle Vaughn Laster
- China-US (Henan) Hormel Cancer Institute, No, 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Jian Li
- China-US (Henan) Hormel Cancer Institute, No, 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Wenna Nie
- China-US (Henan) Hormel Cancer Institute, No, 127 Dongming Road, Zhengzhou, 450008, Henan, China
| | - Yong Weon Yi
- Department of Biochemistry, College of Medicine, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungcheongnam-do, 31116, Republic of Korea
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, 450008, Henan, China
- China-US (Henan) Hormel Cancer Institute, No, 127 Dongming Road, Zhengzhou, 450008, Henan, China
- The Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, 450008, Henan, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, 450008, Henan, China
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungcheongnam-do, 31116, Republic of Korea.
- Graduate School of Convergence Medical Science, Dankook University, Cheonan, Chungcheongnam-do, 31116, Republic of Korea.
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, 450008, Henan, China.
- China-US (Henan) Hormel Cancer Institute, No, 127 Dongming Road, Zhengzhou, 450008, Henan, China.
- The Affiliated Cancer Hospital, Zhengzhou University, Zhengzhou, 450008, Henan, China.
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, 450008, Henan, China.
- International Joint Research Center of Cancer Chemoprevention, Zhengzhou, China.
- The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450008, Henan, China.
| | - Dong Joon Kim
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, 450008, Henan, China.
- China-US (Henan) Hormel Cancer Institute, No, 127 Dongming Road, Zhengzhou, 450008, Henan, China.
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, 450008, Henan, China.
- Department of Microbiology, College of Medicine, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, Chungcheongnam-do, 31116, Republic of Korea.
| |
Collapse
|
39
|
Al-Wahaibi LH, Mohammed AF, Abdelrahman MH, Trembleau L, Youssif BGM. Design, Synthesis, and Biological Evaluation of Indole-2-carboxamides as Potential Multi-Target Antiproliferative Agents. Pharmaceuticals (Basel) 2023; 16:1039. [PMID: 37513950 PMCID: PMC10385579 DOI: 10.3390/ph16071039] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
A small set of indole-based derivatives, IV and Va-I, was designed and synthesized. Compounds Va-i demonstrated promising antiproliferative activity, with GI50 values ranging from 26 nM to 86 nM compared to erlotinib's 33 nM. The most potent antiproliferative derivatives-Va, Ve, Vf, Vg, and Vh-were tested for EGFR inhibitory activity. Compound Va demonstrated the highest inhibitory activity against EGFR with an IC50 value of 71 ± 06 nM, which is higher than the reference erlotinib (IC50 = 80 ± 05 nM). Compounds Va, Ve, Vf, Vg, and Vh were further tested for BRAFV600E inhibitory activity. The tested compounds inhibited BRAFV600E with IC50 values ranging from 77 nM to 107 nM compared to erlotinib's IC50 value of 60 nM. The inhibitory activity of compounds Va, Ve, Vf, Vg, and Vh against VEGFR-2 was also determined. Finally, in silico docking experiments attempted to investigate the binding mode of compounds within the active sites of EGFR, BRAFV600E, and VEGFR-2.
Collapse
Affiliation(s)
- Lamya H Al-Wahaibi
- Department of Chemistry, College of Sciences, Princess Nourah bint Abdulrahman University, Riyadh 11564, Saudi Arabia
| | - Anber F Mohammed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Mostafa H Abdelrahman
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Assiut 71234, Egypt
| | - Laurent Trembleau
- School of Natural and Computing Sciences, University of Aberdeen, Meston Building, Aberdeen AB24 3UE, UK
| | - Bahaa G M Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| |
Collapse
|
40
|
Al-Wabli RI, Issa IS, Al-Mutairi MS, Almomen AA, Attia MI. A Facile Synthesis and Molecular Characterization of Certain New Anti-Proliferative Indole-Based Chemical Entities. Int J Mol Sci 2023; 24:ijms24097862. [PMID: 37175570 PMCID: PMC10178769 DOI: 10.3390/ijms24097862] [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: 03/09/2023] [Revised: 04/06/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Cancer cells frequently develop drug resistance, which leads to chemotherapeutic treatment failure. Additionally, chemotherapies are hindered by their high toxicity. Therefore, the development of new chemotherapeutic drugs with improved clinical outcomes and low toxicity is a major priority. Several indole derivatives exhibit distinctive anti-cancer mechanisms which have been associated with various molecular targets. In this study, target compounds 4a-q were obtained through the reaction of substituted benzyl chloride with hydrazine hydrate, which produces benzyl hydrazine. Subsequently, the appropriate substituted benzyl hydrazine was allowed to react with 1H-indole-2-carboxylic acid or 5-methoxy-1H-indole-2-carboxylic acid using 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide as a coupling agent. All compounds exhibited cytotoxicity in three cell lines, namely, MCF-7, A549, and HCT. Compound 4e exhibited the highest cytotoxicity, with an average IC50 of 2 µM. Moreover, a flow cytometry study revealed a significantly increased prevalence of Annexin-V and 7-AAD positive cell populations. Several derivatives of 4a-q showed moderate to high cytotoxicity against the tested cell lines, with compound 4e having the highest cytotoxicity, indicating that it may possess potential apoptosis-inducing capabilities.
Collapse
Affiliation(s)
- Reem I Al-Wabli
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Iman S Issa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Maha S Al-Mutairi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Aliyah A Almomen
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohamed I Attia
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
41
|
Jacob IT, da Cruz Filho IJ, Alves JEF, de Melo Souza F, de Azevedo RDS, Marques DSC, de Lima Souza TRC, Dos Santos KL, da Rocha Pitta MG, de Melo Rêgo MJB, Oliveira JF, Almeida SMV, do Carmo Alves de Lima M. Interaction study with DNA/HSA, anti-topoisomerase IIα, cytotoxicity and in vitro antiproliferative evaluations and molecular docking of indole-thiosemicarbazone compounds. Int J Biol Macromol 2023; 234:123606. [PMID: 36773880 DOI: 10.1016/j.ijbiomac.2023.123606] [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: 11/15/2022] [Revised: 01/20/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023]
Abstract
In this work we will discuss the antiproliferative evaluation and the possible mechanisms of action of indole-thiosemicarbazone compounds LTs with anti-inflammatory activity, previously described in the literature. In this perspective, some analyzes were carried out, such as the study of binding to human serum albumin (HSA) and to biological targets: DNA and human topoisomerase IIα (topo). Antiproliferative study was performed with DU-145, Jukart, MCF-7 and T-47D tumor lines and J774A.1, besides HepG2 macrophages and hemolytic activity. In the HSA interaction tests, the highest binding constant was 3.70 × 106 M-1, referring to LT89 and in the fluorescence, most compounds, except for LT76 and LT87, promoted fluorescent suppression with the largest Stern-Volmer constant for the LT88 3.55 × 104. In the antiproliferative assay with DU-145 and Jurkat strains, compounds LT76 (0.98 ± 0.10/1.23 ± 0.32 μM), LT77 (0.94 ± 0.05/1.18 ± 0.08 μM) and LT87 (0.94 ± 0.12/0.84 ± 0.09 μM) stood out, due to their IC50 values mentioned above. With the MCF-7 and T-47D cell lines, the lowest IC50 was presented by LT81 with values of 0.74 ± 0.12 μM and 0.68 ± 0.10 μM, respectively, followed by the compounds LT76 and LT87. As well as the positive control amsacrine, the compounds LT76, LT81 and LT87 were able to inhibit the enzymatic action of human Topoisomerase IIα.
Collapse
Affiliation(s)
- Iris Trindade Jacob
- Department of Antibiotics, Federal University of Pernambuco, 50670-901, Brazil
| | | | | | - Felipe de Melo Souza
- Universidade de Pernambuco (UPE), Multicampi Garanhuns, Garanhuns, PE 55290-000, Brazil
| | | | | | | | | | | | | | - Jamerson Ferreira Oliveira
- University for the International Integration of Afro-Brazilian Lusophony (UNILAB), 62790-970 Redenção, CE, Brazil
| | - Sinara Mônica Vitalino Almeida
- Laboratório de Imunopatologia Keizo Asami (LIKA), Universidade Federal de Pernambuco, 50670-901, Brazil; Universidade de Pernambuco (UPE), Multicampi Garanhuns, Garanhuns, PE 55290-000, Brazil.
| | | |
Collapse
|
42
|
Abaev VT, Aksenov NA, Aksenov DA, Aleksandrova EV, Akulova AS, Kurenkov IA, Leontiev AV, Aksenov AV. One-Pot Synthesis of Polynuclear Indole Derivatives by Friedel–Crafts Alkylation of γ-Hydroxybutyrolactams. Molecules 2023; 28:molecules28073162. [PMID: 37049924 PMCID: PMC10095734 DOI: 10.3390/molecules28073162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/22/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
The Friedel–Crafts reaction of novel 3,5-diarylsubstituted 5-hydroxy-1,5-dihydro-2H-pyrrol-2-ones was used for low cost, one-pot preparation of polycyclic indole derivatives structurally similar to Ergot alkaloids.
Collapse
Affiliation(s)
- Vladimir T. Abaev
- Department of Chemistry, Biology and Biotechnology, North-Ossetian State University Named after K. L. Khetagurov, 46 Vatutin St., Vladikavkaz 362025, Russia
- Department of Chemistry, North Caucasus Federal University, 1a Pushkin St., Stavropol 355009, Russia
| | - Nicolai A. Aksenov
- Department of Chemistry, North Caucasus Federal University, 1a Pushkin St., Stavropol 355009, Russia
| | - Dmitrii A. Aksenov
- Department of Chemistry, North Caucasus Federal University, 1a Pushkin St., Stavropol 355009, Russia
| | - Elena V. Aleksandrova
- Department of Chemistry, North Caucasus Federal University, 1a Pushkin St., Stavropol 355009, Russia
| | - Alesia S. Akulova
- Department of Chemistry, North Caucasus Federal University, 1a Pushkin St., Stavropol 355009, Russia
| | - Igor A. Kurenkov
- Department of Chemistry, North Caucasus Federal University, 1a Pushkin St., Stavropol 355009, Russia
| | - Alexander V. Leontiev
- Department of Chemistry, North Caucasus Federal University, 1a Pushkin St., Stavropol 355009, Russia
| | - Alexander V. Aksenov
- Department of Chemistry, North Caucasus Federal University, 1a Pushkin St., Stavropol 355009, Russia
| |
Collapse
|
43
|
Salerno S, Barresi E, Baglini E, Poggetti V, Da Settimo F, Taliani S. Target-Based Anticancer Indole Derivatives for the Development of Anti-Glioblastoma Agents. Molecules 2023; 28:molecules28062587. [PMID: 36985576 PMCID: PMC10056347 DOI: 10.3390/molecules28062587] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive and frequent primary brain tumor, with a poor prognosis and the highest mortality rate. Currently, GBM therapy consists of surgical resection of the tumor, radiotherapy, and adjuvant chemotherapy with temozolomide. Consistently, there are poor treatment options and only modest anticancer efficacy is achieved; therefore, there is still a need for the development of new effective therapies for GBM. Indole is considered one of the most privileged scaffolds in heterocyclic chemistry, so it may serve as an effective probe for the development of new drug candidates against challenging diseases, including GBM. This review analyzes the therapeutic benefit and clinical development of novel indole-based derivatives investigated as promising anti-GBM agents. The existing indole-based compounds which are in the pre-clinical and clinical stages of development against GBM are reported, with particular reference to the most recent advances between 2013 and 2022. The main mechanisms of action underlying their anti-GBM efficacy, such as protein kinase, tubulin and p53 pathway inhibition, are also discussed. The final goal is to pave the way for medicinal chemists in the future design and development of novel effective indole-based anti-GBM agents.
Collapse
|
44
|
El-Zahabi MA, Elkady H, Sakr H, Abdelraheem AS, Eissa SI, El-Adl K. Design, synthesis, anticancer evaluation, in silico docking and ADMET analysis of novel indole-based thalidomide analogs as promising immunomodulatory agents. J Biomol Struct Dyn 2023; 41:15106-15123. [PMID: 36889930 DOI: 10.1080/07391102.2023.2187217] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/23/2023] [Indexed: 03/10/2023]
Abstract
In the present work, novel 16 indole-based thalidomide analogs were designed and synthesized to obtain new effective antitumor immunomodulatory agents. The synthesized compounds were evaluated for their cytotoxic activities against HepG-2, HCT-116, PC3 and MCF-7 cell lines. Generally, the opened analogs of glutarimide ring exhibited higher activities than the closed ones. Compounds 21a-b and 11d,g showed strong potencies against all tested cell lines with IC50 values ranging from 8.27 to 25.20 µM comparable to that of thalidomide (IC50 values ranging from 32.12 to 76.91 µM). The most active compounds were further evaluated for their in vitro immunomodulatory activities via estimation of human tumor necrosis factor alpha (TNF-α), human caspase-8 (CASP8), human vascular endothelial growth factor (VEGF), and nuclear factor kappa-B P65 (NF-κB P65) in HCT-116 cells. Thalidomide was used as a positive control. Compounds 11g, 21a and 21b showed remarkable significant reduction in TNF-α. Furthermore, compounds 11g, 21a and 21b showed significant elevation in CASP8 levels. Compounds 11g and 21a significantly inhibited VEGF. In addition, derivatives 11d, 11g and 21a showed significant decrease in level of NF-κB p65. Moreover, our derivatives exhibited good in silico docking and ADMET profile.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mohamed Ayman El-Zahabi
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Hazem Elkady
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Helmy Sakr
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Adel S Abdelraheem
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Sally I Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Nasser City, Egypt
| | - Khaled El-Adl
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| |
Collapse
|
45
|
Yu C, E R, Zhang XW, Hu WQ, Bao G, Li Y, Liu Y, He Z, Li J, Ma W, Mou LY, Wang R, Sun W. NaClO-Mediated Cross Installation of Indoles and Azoles Benefits Anticancer Hit Discovery. ChemMedChem 2023; 18:e202200651. [PMID: 36585386 DOI: 10.1002/cmdc.202200651] [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] [Received: 11/29/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/01/2023]
Abstract
Innovations in synthetic chemistry have a profound impact on the drug discovery process, and will always be a necessary driver of drug development. As a result, it is of significance to develop novel simple and effective synthetic installation of medicinal modules to promote drug discovery. Herein, we have developed a NaClO-mediated cross installation of indoles and azoles, both of which are frequently encountered in drugs and natural products. This effective toolbox provides a convenient synthetic route to access a library of N-linked 2-(azol-1-yl) indole derivatives, and can be used for late-stage modification of drugs, natural products and peptides. Moreover, biological screening of the library has revealed that several adducts showed promising anticancer activities against A549 and NCI-H1975 cells, which give us a hit for anticancer drug discovery.
Collapse
Affiliation(s)
- Changjun Yu
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Ruiyao E
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Xiao-Wei Zhang
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Wen-Qian Hu
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Guangjun Bao
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Yiping Li
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Yuyang Liu
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Zeyuan He
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Jingyue Li
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Wen Ma
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Ling-Yun Mou
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Rui Wang
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China.,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Wangsheng Sun
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| |
Collapse
|
46
|
Song J, Zhang B, Li M, Zhang J. The current scenario of naturally occurring indole alkaloids with anticancer potential. Fitoterapia 2023; 165:105430. [PMID: 36634875 DOI: 10.1016/j.fitote.2023.105430] [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: 12/18/2022] [Revised: 01/06/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023]
Abstract
Naturally occurring indole alkaloids are ubiquitously present in nature and possess extensive biological properties and structural diversity. Mechanistically, naturally occurring indole alkaloids have the potential to inhibit cancer cell proliferation, arrest cell cycle and induce apoptosis. Accordingly, naturally occurring indole alkaloids exhibit promising activity against both drug-sensitive and drug-resistant cancers including multidrug-resistant forms. Therefore, naturally occurring indole alkaloids constitute an important source of anticancer drug leads and candidates. The goal of this review is to highlight the current scenario of naturally occurring indole alkaloids with anticancer potential, covering articles published from 2018 to present. The names, sources, and antiproliferative activity are discussed to continuously open up a map for the remarkable exploration of more effective candidates.
Collapse
Affiliation(s)
- Juntao Song
- Department of Oncology and Hematology, Zibo 148 Hospital, Zibo 255300, China
| | - Bo Zhang
- Emergency Department, People's Hospital of Zhoucun District, Zibo 255300, China
| | - Ming Li
- Department of Oncology and Hematology, People's Hospital of Zhoucun District, Zibo 255300, China
| | - Jinbiao Zhang
- Department of Oncology and Hematology, Zibo 148 Hospital, Zibo 255300, China.
| |
Collapse
|
47
|
Angelova VT, Tatarova T, Mihaylova R, Vassilev N, Petrov B, Zhivkova Z, Doytchinova I. Novel Arylsulfonylhydrazones as Breast Anticancer Agents Discovered by Quantitative Structure-Activity Relationships. Molecules 2023; 28:2058. [PMID: 36903304 PMCID: PMC10004090 DOI: 10.3390/molecules28052058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Breast cancer (BC) is the second leading cause of cancer death in women, with more than 600,000 deaths annually. Despite the progress that has been made in early diagnosis and treatment of this disease, there is still a significant need for more effective drugs with fewer side effects. In the present study, we derive QSAR models with good predictive ability based on data from the literature and reveal the relationships between the chemical structures of a set of arylsulfonylhydrazones and their anticancer activity on human ER+ breast adenocarcinoma and triple-negative breast (TNBC) adenocarcinoma. Applying the derived knowledge, we design nine novel arylsulfonylhydrazones and screen them in silico for drug likeness. All nine molecules show suitable drug and lead properties. They are synthesized and tested in vitro for anticancer activity on MCF-7 and MDA-MB-231 cell lines. Most of the compounds are more active than predicted and show stronger activity on MCF-7 than on MDA-MB-231. Four of the compounds (1a, 1b, 1c, and 1e) show IC50 values below 1 μM on MCF-7 and one (1e) on MDA-MB-231. The presence of an indole ring bearing 5-Cl, 5-OCH3, or 1-COCH3 has the most pronounced positive effect on the cytotoxic activity of the arylsulfonylhydrazones designed in the present study.
Collapse
Affiliation(s)
| | - Teodora Tatarova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Rositsa Mihaylova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Nikolay Vassilev
- Laboratory “Nuclear Magnetic Resonance”, Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Boris Petrov
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Zvetanka Zhivkova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Irini Doytchinova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| |
Collapse
|
48
|
Al-Wahaibi LH, Mohammed AF, Abdelrahman MH, Trembleau L, Youssif BGM. Design, Synthesis, and Antiproliferative Activity of New 5-Chloro-indole-2-carboxylate and Pyrrolo[3,4- b]indol-3-one Derivatives as Potent Inhibitors of EGFR T790M/BRAF V600E Pathways. Molecules 2023; 28:1269. [PMID: 36770936 PMCID: PMC9921301 DOI: 10.3390/molecules28031269] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Mutant EGFR/BRAF pathways are thought to be crucial targets for the development of anticancer drugs since they are over-activated in several malignancies. We present here the development of a novel series of 5-chloro-indole-2-carboxylate 3a-e, 4a-c and pyrrolo[3,4-b]indol-3-ones 5a-c derivatives as potent inhibitors of mutant EGFR/BRAF pathways with antiproliferative activity. The cell viability assay results of 3a-e, 4a-c, and 5a-c revealed that none of the compounds tested were cytotoxic, and that the majority of those tested at 50 µM had cell viability levels greater than 87%. Compounds 3a-e, 4a-c, and 5a-c had significant antiproliferative activity with GI50 values ranging from 29 nM to 78 nM, with 3a-e outperforming 4a-c and 5a-c in their inhibitory actions against the tested cancer cell lines. Compounds 3a-e were tested for EGFR inhibition, with IC50 values ranging from 68 nM to 89 nM. The most potent derivative was found to be the m-piperidinyl derivative 3e (R = m-piperidin-1-yl), with an IC50 value of 68 nM, which was 1.2-fold more potent than erlotinib (IC50 = 80 nM). Interestingly, all the tested compounds 3a-e had higher anti-BRAFV600E activity than the reference erlotinib but were less potent than vemurafenib, with compound 3e having the most potent activity. Moreover, compounds 3b and 3e showed an 8-fold selectivity index toward EGFRT790M protein over wild-type. Additionally, molecular docking of 3a and 3b against BRAFV600E and EGFRT790M enzymes revealed high binding affinity and active site interactions compared to the co-crystalized ligands. The pharmacokinetics properties (ADME) of 3a-e revealed safety and good pharmacokinetic profile.
Collapse
Affiliation(s)
- Lamya H. Al-Wahaibi
- Department of Chemistry, College of Sciences, Princess Nourah bint Abdulrahman University, Riyadh 11564, Saudi Arabia
| | - Anber F. Mohammed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Mostafa H. Abdelrahman
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Laurent Trembleau
- School of Natural and Computing Sciences, University of Aberdeen, Meston Building, Aberdeen AB24 3UE, UK
| | - Bahaa G. M. Youssif
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| |
Collapse
|
49
|
Rana G, Kar A, Kundal S, Musib D, Jana U. DDQ/Fe(NO 3) 3-Catalyzed Aerobic Synthesis of 3-Acyl Indoles and an In Silico Study for the Binding Affinity of N-Tosyl-3-acyl Indoles toward RdRp against SARS-CoV-2. J Org Chem 2023; 88:838-851. [PMID: 36622749 DOI: 10.1021/acs.joc.2c02009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In the present study, we herein report a DDQ-catalyzed new protocol for the synthesis of substituted 3-acylindoles. Being a potential system for virtual hydrogen storage, introduction of catalytic DDQ in combination with Fe(NO3)3·9H2O and molecular oxygen as co-catalysts offers a regioselective oxo-functionalization of C-3 alkyl-/aryllidine indolines even with scale-up investigations. Intermediate isolation, their spectroscopic characterization, and the density functional theory calculations indicate that the method involves dehydrogenative allylic hydroxylation and 1,3-functional group isomerization/aromatization followed by terminal oxidation to afford 3-acylindoles quantitatively with very high regioselectivity. This method is very general for a large number of substrates with varieties of functional groups tolerance emerging high-yield outcome. Moreover, molecular docking studies were performed for some selected ligands with an RNA-dependent RNA polymerase complex (RdRp complex) of SARS-CoV-2 to illustrate the binding potential of those ligands. The docking results revealed that few of the ligands possess the potential to inhibit the RdRp of SARS-Cov-2 with binding energies (-6.7 to -8.19 kcal/mol), which are comparably higher with respect to the reported binding energies of the conventional re-purposed drugs such as Remdesivir, Ribavirin, and so forth (-4 to -7 kcal/mol).
Collapse
Affiliation(s)
- Gopal Rana
- Department of Chemistry, Jadavpur University, Kolkata 700 032, West Bengal, India
| | - Abhishek Kar
- Department of Chemistry, Jadavpur University, Kolkata 700 032, West Bengal, India
| | - Sandip Kundal
- Department of Chemistry, Jadavpur University, Kolkata 700 032, West Bengal, India
| | - Dulal Musib
- Department of Chemistry, National Institute of Technology Manipur, Langol, Imphal 795004, Manipur, India
| | - Umasish Jana
- Department of Chemistry, Jadavpur University, Kolkata 700 032, West Bengal, India
| |
Collapse
|
50
|
N-Derivatives of ( Z)-Methyl 3-(4-Oxo-2-thioxothiazolidin-5-ylidene)methyl)-1 H-indole-2-carboxylates as Antimicrobial Agents-In Silico and In Vitro Evaluation. Pharmaceuticals (Basel) 2023; 16:ph16010131. [PMID: 36678628 PMCID: PMC9865890 DOI: 10.3390/ph16010131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Herein, we report the experimental evaluation of the antimicrobial activity of seventeen new (Z)-methyl 3-(4-oxo-2-thioxothiazolidin-5-ylidene)methyl)-1H-indole-2-carboxylate derivatives. All tested compounds exhibited antibacterial activity against eight Gram-positive and Gram-negative bacteria. Their activity exceeded those of ampicillin as well as streptomycin by 10-50 fold. The most sensitive bacterium was En. Cloacae, while E. coli was the most resistant one, followed by M. flavus. The most active compound appeared to be compound 8 with MIC at 0.004-0.03 mg/mL and MBC at 0.008-0.06 mg/mL. The antifungal activity of tested compounds was good to excellent with MIC in the range of 0.004-0.06 mg/mL, with compound 15 being the most potent. T. viride was the most sensitive fungal, while A. fumigatus was the most resistant one. Docking studies revealed that the inhibition of E. coli MurB is probably responsible for their antibacterial activity, while 14a-lanosterol demethylase of CYP51Ca is involved in the mechanism of antifungal activity. Furthermore, drug-likeness and ADMET profile prediction were performed. Finally, the cytotoxicity studies were performed for the most active compounds using MTT assay against normal MRC5 cells.
Collapse
|