1
|
Kang Z, Li R, Liu C, Dong X, Hu Y, Xu L, Liu X, Xiang Y, Gao L, Si W, Wang L, Li Q, Zhang L, Wang H, Yang X, Liu J. m 6A-modified cenRNA stabilizes CENPA to ensure centromere integrity in cancer cells. Cell 2024; 187:6035-6054.e27. [PMID: 39305902 DOI: 10.1016/j.cell.2024.08.040] [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: 12/12/2023] [Revised: 01/10/2024] [Accepted: 08/20/2024] [Indexed: 10/20/2024]
Abstract
m6A modification is best known for its critical role in controlling multiple post-transcriptional processes of the mRNAs. Here, we discovered elevated levels of m6A modification on centromeric RNA (cenRNA) in cancerous cells compared with non-cancerous cells. We then identified CENPA, an H3 variant, as an m6A reader of cenRNA. CENPA is localized at centromeres and is essential in preserving centromere integrity and function during mitosis. The m6A-modified cenRNA stabilizes centromeric localization of CENPA in cancer cells during the S phase of the cell cycle. Mutations of CENPA at the Leu61 and the Arg63 or removal of cenRNA m6A modification lead to loss of centromere-bound CENPA during S phase. This in turn results in compromised centromere integrity and abnormal chromosome separation and hinders cancer cell proliferation and tumor growth. Our findings unveil an m6A reading mechanism by CENPA that epigenetically governs centromere integrity in cancer cells, providing potential targets for cancer therapy.
Collapse
Affiliation(s)
- Zihong Kang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China
| | - Ruimeng Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Science, Tsinghua University, 100084 Beijing, China
| | - Chang Liu
- Department of Genetics, Stanford University, School of Medicine, Stanford, CA, USA
| | - Xiaozhe Dong
- College of Chemistry and Molecular Engineering, Peking University, 100871 Beijing, China
| | - Yuxuan Hu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 211198 Nanjing, China
| | - Lei Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008 Nanjing, China
| | - Xinyu Liu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China
| | - Yunfan Xiang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China
| | - Liming Gao
- School of Science, China Pharmaceutical University, 211198 Nanjing, China
| | - Wenzhe Si
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Laboratory Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Lei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008 Nanjing, China
| | - Qing Li
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China
| | - Liang Zhang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 310022 Hangzhou, China
| | - Huan Wang
- College of Chemistry and Molecular Engineering, Peking University, 100871 Beijing, China
| | - Xuerui Yang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Science, Tsinghua University, 100084 Beijing, China.
| | - Jun Liu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China.
| |
Collapse
|
2
|
Eigenfeld M, Lupp KFM, Schwaminger SP. Role of Natural Binding Proteins in Therapy and Diagnostics. Life (Basel) 2024; 14:630. [PMID: 38792650 PMCID: PMC11122601 DOI: 10.3390/life14050630] [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: 03/31/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
This review systematically investigates the critical role of natural binding proteins (NBPs), encompassing DNA-, RNA-, carbohydrate-, fatty acid-, and chitin-binding proteins, in the realms of oncology and diagnostics. In an era where cancer continues to pose significant challenges to healthcare systems worldwide, the innovative exploration of NBPs offers a promising frontier for advancing both the diagnostic accuracy and therapeutic efficacy of cancer management strategies. This manuscript provides an in-depth examination of the unique mechanisms by which NBPs interact with specific molecular targets, highlighting their potential to revolutionize cancer diagnostics and therapy. Furthermore, it discusses the burgeoning research on aptamers, demonstrating their utility as 'nucleic acid antibodies' for targeted therapy and precision diagnostics. Despite the promising applications of NBPs and aptamers in enhancing early cancer detection and developing personalized treatment protocols, this review identifies a critical knowledge gap: the need for comprehensive studies to understand the diverse functionalities and therapeutic potentials of NBPs across different cancer types and diagnostic scenarios. By bridging this gap, this manuscript underscores the importance of NBPs and aptamers in paving the way for next-generation diagnostics and targeted cancer treatments.
Collapse
Affiliation(s)
- Marco Eigenfeld
- Otto-Loewi Research Center, Division of Medicinal Chemistry, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Kilian F. M. Lupp
- Otto-Loewi Research Center, Division of Medicinal Chemistry, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Sebastian P. Schwaminger
- Otto-Loewi Research Center, Division of Medicinal Chemistry, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
3
|
Long A, Oswood CJ, Kelly CB, Bryan MC, MacMillan DWC. Couple-close construction of polycyclic rings from diradicals. Nature 2024; 628:326-332. [PMID: 38480891 PMCID: PMC11487475 DOI: 10.1038/s41586-024-07181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/08/2024] [Indexed: 04/06/2024]
Abstract
Heteroarenes are ubiquitous motifs in bioactive molecules, conferring favourable physical properties when compared to their arene counterparts1-3. In particular, semisaturated heteroarenes possess attractive solubility properties and a higher fraction of sp3 carbons, which can improve binding affinity and specificity. However, these desirable structures remain rare owing to limitations in current synthetic methods4-6. Indeed, semisaturated heterocycles are laboriously prepared by means of non-modular fit-for-purpose syntheses, which decrease throughput, limit chemical diversity and preclude their inclusion in many hit-to-lead campaigns7-10. Herein, we describe a more intuitive and modular couple-close approach to build semisaturated ring systems from dual radical precursors. This platform merges metallaphotoredox C(sp2)-C(sp3) cross-coupling with intramolecular Minisci-type radical cyclization to fuse abundant heteroaryl halides with simple bifunctional feedstocks, which serve as the diradical synthons, to rapidly assemble a variety of spirocyclic, bridged and substituted saturated ring types that would be extremely difficult to make by conventional methods. The broad availability of the requisite feedstock materials allows sampling of regions of underexplored chemical space. Reagent-controlled radical generation leads to a highly regioselective and stereospecific annulation that can be used for the late-stage functionalization of pharmaceutical scaffolds, replacing lengthy de novo syntheses.
Collapse
Affiliation(s)
- Alice Long
- Merck Center for Catalysis at Princeton University, Princeton, NJ, USA
| | | | - Christopher B Kelly
- Discovery Process Research, Janssen Research and Development LLC, Spring House, PA, USA
| | - Marian C Bryan
- Therapeutics Discovery, Janssen Research and Development LLC, Spring House, PA, USA
| | | |
Collapse
|
4
|
Rząd K, Gabriel I, Paluszkiewicz E, Kuplińska A, Olszewski M, Chylewska A, Dąbrowska AM, Kozłowska-Tylingo K. Targeting yeast topoisomerase II by imidazo and triazoloacridinone derivatives resulting in their antifungal activity. Sci Rep 2024; 14:3594. [PMID: 38351313 PMCID: PMC10864382 DOI: 10.1038/s41598-024-54252-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/10/2024] [Indexed: 02/16/2024] Open
Abstract
Fungal pathogens are considered as serious factors for deadly diseases and are a case of medical concern. Invasive fungal infections also complicate the clinical course of COVID-19, leading to a significant increase in mortality. Furthermore, fungal strains' multidrug resistance has increased the demand for antifungals with a different mechanism of action. The present study aimed to identify antifungal compounds targeting yeast topoisomerase II (yTOPOII) derived from well-known human topoisomerase II (hTOPOII) poisons C-1305 and C-1311. Two sets of derivatives: triazoloacridinones (IKE1-8) and imidazoacridinones (IKE9-14) were synthetized and evaluated with a specific emphasis on the molecular mechanism of action. Our results indicated that their effectiveness as enzyme inhibitors was not solely due to intercalation ability but also as a result of influence on catalytic activity by the formation of covalent complexes between plasmid DNA and yTOPOII. Lysine conjunction increased the strength of the compound's interaction with DNA and improved penetration into the fungal cells. Triazoloacridinone derivatives in contrast to starting compound C-1305 exhibited moderate antifungal activity and at least twice lower cytotoxicity. Importantly, compounds (IKE5-8) were not substrates for multidrug ABC transporters whereas a derivative conjugated with lysine (IKE7), showed the ability to overcome C. glabrata fluconazole-resistance (MIC 32-64 µg mL-1).
Collapse
Affiliation(s)
- Kamila Rząd
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry and BioTechMed Center, Gdansk University of Technology, 11/12 Narutowicza Str., 80-233, Gdansk, Poland.
| | - Iwona Gabriel
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry and BioTechMed Center, Gdansk University of Technology, 11/12 Narutowicza Str., 80-233, Gdansk, Poland
| | - Ewa Paluszkiewicz
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry and BioTechMed Center, Gdansk University of Technology, 11/12 Narutowicza Str., 80-233, Gdansk, Poland
| | - Aleksandra Kuplińska
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry and BioTechMed Center, Gdansk University of Technology, 11/12 Narutowicza Str., 80-233, Gdansk, Poland
| | - Mateusz Olszewski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry and BioTechMed Center, Gdansk University of Technology, 11/12 Narutowicza Str., 80-233, Gdansk, Poland
| | - Agnieszka Chylewska
- Department of Bioinorganic Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Aleksandra M Dąbrowska
- Department of Bioinorganic Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Katarzyna Kozłowska-Tylingo
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry and BioTechMed Center, Gdansk University of Technology, 11/12 Narutowicza Str., 80-233, Gdansk, Poland
| |
Collapse
|
5
|
Dar A, Godara P, Prusty D, Bashir M. Plasmodium falciparum topoisomerases: Emerging targets for anti-malarial therapy. Eur J Med Chem 2024; 265:116056. [PMID: 38171145 DOI: 10.1016/j.ejmech.2023.116056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024]
Abstract
Different metabolic pathways like DNA replication, transcription, and recombination generate topological constrains in the genome. These topological constraints are resolved by essential molecular machines known as topoisomerases. To bring changes in DNA topology, the topoisomerases create a single or double-stranded nick in the template DNA, hold the nicked ends to let the tangled DNA pass through, and finally re-ligate the breaks. The DNA nicking and re-ligation activities as well as ATPase activities (when present) in topoisomerases are subjected to inhibition by several anticancer and antibacterial drugs, thus establishing these enzymes as successful targets in anticancer and antibacterial therapies. The anti-topoisomerase drugs interfere with the functioning of these enzymes and result in the accumulation of DNA tangles or lethal genomic breaks, thereby promoting host cell (or organism) death. The potential of topoisomerases in the human malarial parasite, Plasmodium falciparum in antimalarial drug development has received little attention so far. Interestingly, the parasite genome encodes orthologs of topoisomerases found in eukaryotes, prokaryotes, and archaea, thus, providing an enormous opportunity for investigating these enzymes for antimalarial therapeutics. This review focuses on the features of Plasmodium falciparum topoisomerases (PfTopos) with respect to their closer counterparts in other organisms. We will discuss overall advances and basic challenges with topoisomerase research in Plasmodium falciparum and our attempts to understand the interaction of PfTopos with classical and new-generation topoisomerase inhibitors using in silico molecular docking approach. The recent episodes of parasite resistance against artemisinin, the only effective antimalarial drug at present, further highlight the significance of investigating new drug targets including topoisomerases in antimalarial therapeutics.
Collapse
Affiliation(s)
- Ashraf Dar
- Department of Biochemistry, University of Kashmir, Srinagar, 190006, India.
| | - Priya Godara
- Central University of Rajasthan, Ajmer, Rajasthan, India
| | | | - Masarat Bashir
- COTS, Sheri-Kashmir University of Agricultural Sciences and Technology, Mirgund, Srinagar, India
| |
Collapse
|
6
|
Peng X, Wu H, Zhang B, Xu C, Lang J. A Novel Nucleic Acid Sensing-related Genes Signature for Predicting Immunotherapy Efficacy and Prognosis of Lung Adenocarcinoma. Curr Cancer Drug Targets 2024; 24:425-444. [PMID: 37592781 DOI: 10.2174/1568009623666230817101843] [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: 03/22/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND As a novel pillar for lung adenocarcinoma (LUAD) treatment, immunotherapy has limited efficiency in LUAD patients. The nucleic acid sensing (NAS) pathways are critical in the anti-tumor immune response, but their role in LUAD remains controversial. OBJECTIVE The study aims to develop a classification system to identify immune subtypes of LUAD based on nucleic acid sensing-related genes so that it can help screen patients who may respond to immunotherapy. METHODS We performed a comprehensive bioinformatics analysis of the NAS molecule expression profiles across multiple public datasets. Using qRT-PCR to verify the NAS genes in multiple lung cancer cell lines. Molecular docking was performed to screen drug candidates. RESULTS The NAS-activated subgroup and NAS-suppressed subgroup were validated based on the different patterns of gene expression and pathways enrichment. The NAS-activated subgroup displayed a stronger immune infiltration and better prognosis of patients. Moreover, we constructed a seven nucleic acid sensing-related risk score (NASRS) model for the convenience of clinical application. The predictive values of NASRS in prognosis and immunotherapy were subsequently fully validated in the lung adenocarcinoma dataset and the uroepithelial carcinoma dataset. Additionally, five potential drugs binding to the core target of the NAS signature were predicted through molecular docking. CONCLUSION We found a significant correlation between nucleic acid sensing function and the immune treatment efficiency in LUAD. The NASRS can be used as a robust biomarker for the predicting of prognosis and immunotherapy efficiency and may help in clinical decisions for LUAD patients.
Collapse
Affiliation(s)
- Xinhao Peng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Hong Wu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Biqin Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Chuan Xu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jinyi Lang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
7
|
Dong H, Lu L, Song X, Li Y, Zhou J, Xu Y, Zhang Y, Qi J, Liang T, Wang J. Design, synthesis and biological evaluation of tetrahydroquinoxaline sulfonamide derivatives as colchicine binding site inhibitors. RSC Adv 2023; 13:30202-30216. [PMID: 37849704 PMCID: PMC10577396 DOI: 10.1039/d3ra05720h] [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: 08/22/2023] [Accepted: 10/09/2023] [Indexed: 10/19/2023] Open
Abstract
Colchicine binding site inhibitors (CBSIs) are potential microtubule targeting agents (MTAs), which can overcome multidrug resistance, improve aqueous solubility and reduce toxicity faced by most MTAs. Novel tetrahydroquinoxaline sulfonamide derivatives were designed, synthesized and evaluated for their antiproliferative activities. The MTT assay results demonstrated that some derivatives exhibited moderate to strong inhibitory activities against HT-29 cell line. Among them, compound I-7 was the most active compound. Moreover, I-7 inhibited tubulin polymerization, disturbed microtubule network, disrupted the formation of mitotic spindle and arrested cell cycle at G2/M phase. However, I-7 didn't induce cell apoptosis. Furthermore, the prediction of ADME demonstrated that I-7 showed favorable physiochemical and pharmacokinetic properties. And the detailed molecular docking confirmed I-7 targeted the site of colchicine through hydrogen and hydrophobic interactions.
Collapse
Affiliation(s)
- Haiyang Dong
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Lu Lu
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Xueting Song
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Youkang Li
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Jinguang Zhou
- Huaihe Hospital of Henan University Kaifeng 475004 Henan China
| | - Yungen Xu
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 211198 China
| | - Yahong Zhang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Jianguo Qi
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Tingting Liang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Jianhong Wang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| |
Collapse
|
8
|
Lee YC, Chiou JT, Wang LJ, Chen YJ, Chang LS. Amsacrine downregulates BCL2L1 expression and triggers apoptosis in human chronic myeloid leukemia cells through the SIDT2/NOX4/ERK/HuR pathway. Toxicol Appl Pharmacol 2023; 474:116625. [PMID: 37451322 DOI: 10.1016/j.taap.2023.116625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Accumulating evidence indicates that the anticancer activity of acridine derivatives is mediated through the regulation of anti-apoptotic and pro-apoptotic BCL2 protein expression. Therefore, we investigated whether the cytotoxicity of amsacrine with an acridine structural scaffold in human chronic myeloid leukemia (CML) K562 cells was mediated by BCL2 family proteins. Amsacrine induced apoptosis, mitochondrial depolarization, and BCL2L1 (also known as BCL-XL) downregulation in K562 cells. BCL2L1 overexpression inhibited amsacrine-induced cell death and mitochondrial depolarization. Amsacrine treatment triggered SIDT2-mediated miR-25 downregulation, leading to increased NOX4-mediated ROS production. ROS-mediated inactivation of ERK triggered miR-22 expression, leading to increased HuR mRNA decay. As HuR is involved in stabilizing BCL2L1 mRNA, downregulation of BCL2L1 was noted in K562 cells after amsacrine treatment. In contrast, amsacrine-induced BCL2L1 downregulation was alleviated by restoring ERK phosphorylation and HuR expression. Altogether, the results of this study suggest that amsacrine triggers apoptosis in K562 cells by inhibiting BCL2L1 expression through the SIDT2/NOX4/ERK-mediated downregulation of HuR. Furthermore, a similar pathway also explains the cytotoxicity of amsacrine in CML MEG-01 and KU812 cells.
Collapse
Affiliation(s)
- Yuan-Chin Lee
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Jing-Ting Chiou
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Liang-Jun Wang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Ying-Jung Chen
- Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
9
|
Abdullah S, Ganguly S. An overview of imidazole and its analogues as potent anticancer agents. Future Med Chem 2023; 15:1621-1646. [PMID: 37727960 DOI: 10.4155/fmc-2023-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023] Open
Abstract
The quest for novel, physiologically active imidazoles remains an exciting topic of research among medicinal chemists. The imidazole ring is a five-membered aromatic heterocycle that is found in both natural and synthesized compounds. Multiple anticancer drug classes are currently available on the market, but concerns including toxicity, limited efficacy and solubility have lowered the overall therapeutic index. Therefore, the hunt for new potential chemotherapeutic agents persists. The development of imidazole as a reliable and safer alternative to anticancer treatment is generating much attention among experts. Tubulin or microtubule polymerization inhibition and changes in the structure and function of DNA, VEGF, topoisomerase, kinases, histone deacetylases and certain other proteins that affect gene expression are among the putative targets.
Collapse
Affiliation(s)
- Salik Abdullah
- Department of Pharmaceutical Sciences, Birla Institute of Technology, Mesra, Jharkhand, 835215, India
| | - Swastika Ganguly
- Department of Pharmaceutical Sciences, Birla Institute of Technology, Mesra, Jharkhand, 835215, India
| |
Collapse
|
10
|
Elanany MA, Osman EEA, Gedawy EM, Abou-Seri SM. Design and synthesis of novel cytotoxic fluoroquinolone analogs through topoisomerase inhibition, cell cycle arrest, and apoptosis. Sci Rep 2023; 13:4144. [PMID: 36914702 PMCID: PMC10011602 DOI: 10.1038/s41598-023-30885-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/02/2023] [Indexed: 03/16/2023] Open
Abstract
To exploit the advantageous properties of approved drugs to hasten anticancer drug discovery, we designed and synthesized a series of fluoroquinolone (FQ) analogs via functionalization of the acid hydrazides of moxifloxacin, ofloxacin, and ciprofloxacin. Under the NCI-60 Human Tumor Cell Line Screening Assay, (IIIf) was the most potent among moxifloxacin derivatives, whereas (VIb) was the only ofloxacin derivative with significant effects and ciprofloxacin derivatives were devoid of activity. (IIIf) and (VIb) were further selected for five-dose evaluation, where they showed potent growth inhibition with a mean GI50 of 1.78 and 1.45 µM, respectively. (VIb) elicited a more potent effect reaching sub-micromolar level on many cell lines, including MDA-MB-468 and MCF-7 breast cancer cell lines (GI50 = 0.41 and 0.42 µM, respectively), NSCLC cell line HOP-92 (GI50 = 0.50 µM) and CNS cell lines SNB-19 and U-251 (GI50 = 0.51 and 0.61 µM, respectively). (IIIf) and (VIb) arrested MCF-7 cells at G1/S and G1, respectively, and induced apoptosis mainly through the intrinsic pathway as shown by the increased ratio of Bax/Bcl-2 and caspase-9 with a lesser activation of the extrinsic pathway through caspase-8. Both compounds inhibited topoisomerase (Topo) with preferential activity on type II over type I and (VIb) was marginally more potent than (IIIf). Docking study suggests that (IIIf) and (VIb) bind differently to Topo II compared to etoposide. (IIIf) and (VIb) possess high potential for oral absorption, low CNS permeability and low binding to plasma proteins as suggested by in silico ADME calculations. Collectively, (IIIf) and (VIb) represent excellent lead molecules for the development of cytotoxic agents from quinolone scaffolds.
Collapse
Affiliation(s)
- Mohamed A Elanany
- Department of Pharmaceutical Chemistry, School of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.
| | - Essam Eldin A Osman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ehab Mohamed Gedawy
- Department of Pharmaceutical Chemistry, School of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.,Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Sahar M Abou-Seri
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
11
|
Krochtová K, Halečková A, Janovec L, Blizniaková M, Kušnírová K, Kožurková M. Novel 3,9-Disubstituted Acridines with Strong Inhibition Activity against Topoisomerase I: Synthesis, Biological Evaluation and Molecular Docking Study. Molecules 2023; 28:1308. [PMID: 36770975 PMCID: PMC9921529 DOI: 10.3390/molecules28031308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
A series of novel 3,9-disubstituted acridines were synthesized and their biological potential was investigated. The synthetic plan consists of eight reaction steps, which produce the final products, derivatives 17a-17j, in a moderate yield. The principles of cheminformatics and computational chemistry were applied in order to study the relationship between the physicochemical properties of the 3,9-disubstituted acridines and their biological activity at a cellular and molecular level. The selected 3,9-disubstituted acridine derivatives were studied in the presence of DNA using spectroscopic (UV-Vis, circular dichroism, and thermal denaturation) and electrophoretic (nuclease activity, relaxation and unwinding assays for topoisomerase I and decatenation assay for topoisomerase IIα) methods. Binding constants (2.81-9.03 × 104 M-1) were calculated for the derivatives from the results of the absorption titration spectra. The derivatives were found to have caused the inhibition of both topoisomerase I and topoisomerase IIα. Molecular docking simulations suggested a different way in which the acridines 17a-17j can interact with topoisomerase I versus topoisomerase IIα. A strong correlation between the lipophilicity of the derivatives and their ability to stabilize the intercalation complex was identified for all of the studied agents. Acridines 17a-17j were also subjected to in vitro screening conducted by the Developmental Therapeutic Program of the National Cancer Institute (NCI) against a panel of 60 cancer cell lines. The strongest biological activity was displayed by aniline acridine 17a (MCF7-GI50 18.6 nM) and N,N-dimethylaniline acridine 17b (SR-GI50 38.0 nM). The relationship between the cytostatic activity of the most active substances (derivatives 17a, 17b, and 17e-17h) and their values of KB, LogP, ΔS°, and δ was also investigated. Due to the fact that a significant correlation was only found in the case of charge density, δ, it is possible to assume that the cytostatic effect might be dependent upon the structural specificity of the acridine derivatives.
Collapse
Affiliation(s)
- Kristína Krochtová
- Department of Biochemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Moyzesova 11, 040 01 Košice, Slovakia
| | - Annamária Halečková
- Department of Organic Chemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Moyzesova 11, 040 01 Košice, Slovakia
| | - Ladislav Janovec
- Department of Organic Chemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Moyzesova 11, 040 01 Košice, Slovakia
| | - Michaela Blizniaková
- Department of Biochemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Moyzesova 11, 040 01 Košice, Slovakia
| | - Katarína Kušnírová
- Department of Organic Chemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Moyzesova 11, 040 01 Košice, Slovakia
| | - Mária Kožurková
- Department of Biochemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Moyzesova 11, 040 01 Košice, Slovakia
| |
Collapse
|
12
|
Okoro CO, Fatoki TH. A Mini Review of Novel Topoisomerase II Inhibitors as Future Anticancer Agents. Int J Mol Sci 2023; 24:ijms24032532. [PMID: 36768852 PMCID: PMC9916523 DOI: 10.3390/ijms24032532] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
Several reviews of inhibitors of topoisomerase II have been published, covering research before 2018. Therefore, this review is focused primarily on more recent publications with relevant points from the earlier literature. Topoisomerase II is an established target for anticancer drugs, which are further subdivided into poisons and catalytic inhibitors. While most of the topoisomerase II-based drugs in clinical use are mostly topoisomerase II poisons, their mechanism of action has posed severe concern due to DNA damaging potential, including the development of multi-drug resistance. As a result, we are beginning to see a gradual paradigm shift towards non-DNA damaging agents, such as the lesser studied topoisomerase II catalytic inhibitors. In addition, this review describes some novel selective catalytic topoisomerase II inhibitors. The ultimate goal is to bring researchers up to speed by curating and delineating new scaffolds as the leads for the optimization and development of new potent, safe, and selective agents for the treatment of cancer.
Collapse
|
13
|
Synthesis and Evaluation of Antiproliferative Activity, Topoisomerase IIα Inhibition, DNA Binding and Non-Clinical Toxicity of New Acridine-Thiosemicarbazone Derivatives. Pharmaceuticals (Basel) 2022; 15:ph15091098. [PMID: 36145320 PMCID: PMC9506480 DOI: 10.3390/ph15091098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/16/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, we report the synthesis of twenty new acridine–thiosemicarbazone derivatives and their antiproliferative activities. Mechanisms of action such as the inhibition of topoisomerase IIα and the interaction with DNA have been studied for some of the most active derivatives by means of both in silico and in vitro methods, and evaluations of the non-clinical toxicities (in vivo) in mice. In general, the compounds showed greater cytotoxicity against B16-F10 cells, with the highest potency for DL-08 (IC50 = 14.79 µM). Derivatives DL-01 (77%), DL-07 (74%) and DL-08 (79%) showed interesting inhibition of topoisomerase IIα when compared to amsacrine, at 100 µM. In silico studies proposed the way of bonding of these compounds and a possible stereoelectronic reason for the absence of enzymatic activity for CL-07 and DL-06. Interactions with DNA presented different spectroscopic effects and indicate that the compound CL-07 has higher affinity for DNA (Kb = 4.75 × 104 M−1; Ksv = 2.6 × 103 M−1). In addition, compounds selected for non-clinical toxicity testing did not show serious signs of toxicity at the dose of 2000 mg/kg in mice; cytotoxic tests performed on leukemic cells (K-562) and its resistant form (K-562 Lucena 1) identified moderate potency for DL-01 and DL-08, with IC50 between 11.45 and 17.32 µM.
Collapse
|
14
|
Prostate Apoptosis Response-4 (Par-4): A Novel Target in Pyronaridine-Induced Apoptosis in Glioblastoma (GBM) Cells. Cancers (Basel) 2022; 14:cancers14133198. [PMID: 35804970 PMCID: PMC9264948 DOI: 10.3390/cancers14133198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary GBM treatment is an area of high unmet need due to the heterogeneous and anaplastic character of this cancer in turn leading to an extremely poor prognosis. Finding new molecular entities by traditional or de novo approaches to drug discovery is lengthy and expensive. Repurposing existing drugs can be attractive as the process is often less risky, more cost, and time-effective. Amongst potential drug-repurposing candidates, Pyronaridine (PYR), an antimalarial drug has shown anti-cancer effects against several cancers, however, its potential for the treatment of GBM has not been explored. In this study, we have identified a unique mechanism of action of PYR against GBM by upregulating a tumor suppressor protein, Par-4 along with the elucidation of the complex network of pathways mediated through Par-4 leading to GBM cell death. Abstract Glioblastoma (GBM) is an aggressive form of brain tumor with a median survival of approximately 12 months. With no new drugs in the last few decades and limited success in clinics for known therapies, drug repurposing is an attractive choice for its treatment. Here, we examined the efficacy of pyronaridine (PYR), an anti-malarial drug in GBM cells. PYR induced anti-proliferative activity in GBM cells with IC50 ranging from 1.16 to 6.82 µM. Synergistic activity was observed when PYR was combined with Doxorubicin and Ritonavir. Mechanistically, PYR triggered mitochondrial membrane depolarization and enhanced the ROS levels causing caspase-3 mediated apoptosis. PYR significantly decreased markers associated with proliferation, EMT, hypoxia, and stemness and upregulated the expression of E-cadherin. Interestingly, PYR induced the expression of intracellular as well as secretory Par-4, a tumor suppressor in GBM cells, which was confirmed using siRNA. Notably, Par-4 levels in plasma samples of GBM patients were significantly lower than normal healthy volunteers. Thus, our study demonstrates for the first time that PYR can be repurposed against GBM with a novel mechanism of action involving Par-4. Herewith, we discuss the role of upregulated Par-4 in a highly interconnected signaling network thereby advocating its importance as a therapeutic target.
Collapse
|
15
|
Sing LC, Roy A, Hui LY, Mun CS, Rajak H, Karunakaran R, Ravichandran V. Multi-targeted molecular docking, drug-likeness and ADMET studies of derivatives of few quinoline- and acridine-based FDA-approved drugs for anti-breast cancer activity. Struct Chem 2022. [DOI: 10.1007/s11224-022-01878-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
16
|
Bahrami A, Makiabadi E, Jalali S, Heidari Z, Assadi M, Rashidkhani B. Dietary Intake of Polyphenols and the Risk of Breast Cancer: a Case-Control Study. Clin Nutr Res 2021; 10:330-340. [PMID: 34796137 PMCID: PMC8575644 DOI: 10.7762/cnr.2021.10.4.330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 11/30/2022] Open
Abstract
Despite mounting evidence that dietary polyphenols might have a protective role against the risk of breast cancer (BC), few studies have assessed the relationship between intake of polyphenol classes and subclasses with BC. Thus, we examined the relationship between dietary polyphenol classes and individual polyphenol subclasses and the risk of BC. Overall, 134 newly diagnosed BC patients and 267 healthy hospitalized controls were studied. Dietary intake was assessed using a validated 168-item food frequency questionnaire (FFQ). To estimate dietary intake of polyphenols, polyphenol content (flavonoids, lignans, stilbenes and phenolic acids) of 80 food items were derived from an updated version of the phenol explorer database containing information on the effects of food processing on polyphenol content. The dietary polyphenol intake was calculated by matching the subjects' food consumption data with our polyphenol content database. Multivariate logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). Controls had higher intake of total polyphenol (marginally significant; p = 0.07), hydroxycinnamic acid (marginally significant; p = 0.05) and lignan (p = 0.01). After adjusting for potential confounders, high consumption of lignans (highest vs. lowest tertile: OR, 0.51; 95% CI, 0.26–0.97; p for trend = 0.04) associated with decreased risk of BC. There was no significant relationship between intake of other polyphenols and risk of BC. Our findings suggest that high lignan intake is associated with a reduced risk of BC.
Collapse
Affiliation(s)
- Alireza Bahrami
- Student Research Committee, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran 1981619573, Iran.,Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran 1981619573, Iran
| | - Elham Makiabadi
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran 1981619573, Iran
| | - Saba Jalali
- Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran 1981619573, Iran
| | - Zeinab Heidari
- Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran 1981619573, Iran
| | - Mojan Assadi
- Department of Oncology, Shahid Madani Hospital, Alborz University of Medical Science, Karaj 3149779453, Iran
| | - Bahram Rashidkhani
- Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran 1981619573, Iran
| |
Collapse
|
17
|
Kozurkova M. Acridine derivatives as inhibitors/poisons of topoisomerase II. J Appl Toxicol 2021; 42:544-552. [PMID: 34514603 DOI: 10.1002/jat.4238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 12/18/2022]
Abstract
The potential of acridines (amsacrine) as a topoisomerase II inhibitor or poison was first discovered in 1984, and since then, a considerable number of acridine derivatives have been tested as topoisomerase inhibitors/poisons, containing different substituents on the acridine chromophore. This review will discuss a series of studies published over the course of the last decade, which have investigated various novel acridine derivatives against topoisomerase II activity.
Collapse
Affiliation(s)
- Maria Kozurkova
- Department of Biochemistry, Institute of Chemistry, Faculty of Science, P. J. Šafárik University, Kosice, Slovak Republic.,Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
18
|
Lauria A, La Monica G, Bono A, Martorana A. Quinoline anticancer agents active on DNA and DNA-interacting proteins: From classical to emerging therapeutic targets. Eur J Med Chem 2021; 220:113555. [PMID: 34052677 DOI: 10.1016/j.ejmech.2021.113555] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 12/25/2022]
Abstract
Quinoline is one of the most important and versatile nitrogen heterocycles embodied in several biologically active molecules. Within the numerous quinolines developed as antiproliferative agents, this review is focused on compounds interfering with DNA structure or with proteins/enzymes involved in the regulation of double helix functional processes. In this light, a special focus is given to the quinoline compounds, acting with classical/well-known mechanisms of action (DNA intercalators or Topoisomerase inhibitors). In particular, the quinoline drugs amsacrine and camptothecin (CPT) have been studied as key lead compounds for the development of new agents with improved PK and tolerability properties. Moreover, notable attention has been paid to the quinoline molecules, which are able to interfere with emerging targets involved in cancer progression, as G-quadruplexes or the epigenetic ones (e.g.: histone deacetylase, DNA and histones methyltransferase). The antiproliferative and the enzymatic inhibition data of the reviewed compounds have been analyzed. Furthermore, concerning the SAR (structure-activity relationship) aspects, the most recurrent ligand-protein interactions are summarized, underling the structural requirements for each kind of mechanism of action.
Collapse
Affiliation(s)
- Antonino Lauria
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy
| | - Gabriele La Monica
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy
| | - Alessia Bono
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy
| | - Annamaria Martorana
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy.
| |
Collapse
|
19
|
Man RJ, Jeelani N, Zhou C, Yang YS. Recent Progress in the Development of Quinoline Derivatives for the Exploitation of Anti-Cancer Agents. Anticancer Agents Med Chem 2021; 21:825-838. [PMID: 32416703 DOI: 10.2174/1871520620666200516150345] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/23/2020] [Accepted: 02/10/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Along with the progress in medicine and therapies, the exploitation of anti-cancer agents focused more on the vital signaling pathways and key biological macromolecules. With rational design and advanced synthesis, quinoline derivatives have been utilized frequently in medicinal chemistry, especially in developing anti-cancer drugs or candidates. METHODS Using DOI searching, articles published before 2020 all over the world have been reviewed as comprehensively as possible. RESULTS In this review, we selected the representative quinoline derivate drugs in market or clinical trials, classified them into five major categories with detailed targets according to their main mechanisms, discussed the relationship within the same mechanism, and generated a summative discussion with prospective expectations. For each mechanism, the introduction of the target was presented, with the typical examples of quinoline derivate drugs. CONCLUSION This review has highlighted the quinoline drugs or candidates, suited them into corresponding targets in their pathways, summarized and discussed. We hope that this review may help the researchers who are interested in discovering quinoline derivate anti-cancer agents obtain considerable understanding of this specific topic. Through the flourishing period and the vigorous strategies in clinical trials, quinoline drugs would be potential but facing new challenges in the future.
Collapse
Affiliation(s)
- Ruo-Jun Man
- College of Chemistry and Chemical Engineering, Guangxi University for Nationalities, Nanning, China
| | - Nasreen Jeelani
- Institute of Chemistry and BioMedical Sciences, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Chongchen Zhou
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, 450018, China
| | - Yu-Shun Yang
- Institute of Chemistry and BioMedical Sciences, School of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
20
|
Ayyamperumal S, DJ D, Tallapaneni V, Mohan S, S B, Selvaraj J, Joghee NM, MJN C. Molecular docking analysis of α-Topoisomerase II with δ-Carboline derivatives as potential anticancer agents. Bioinformation 2021; 17:249-265. [PMID: 34393444 PMCID: PMC8340707 DOI: 10.6026/97320630017249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/30/2021] [Accepted: 01/31/2021] [Indexed: 11/29/2022] Open
Abstract
The enzyme, α-topoisomerase II (α-Topo II), is known to regulate efficiently the topology of DNA. It is highly expressed in rapidly proliferating cells and plays an important role in replication, transcription and chromosome organisation. This has prompted several investigators to pursue α-Topo II inhibitors as anticancer agents. δ-Carboline, a natural product, and its synthetic derivatives are known to exert potent anticancer activity by selectively targeting α-Topo II. Therefore, it is of interest to design carboline derivatives fused with pyrrolidine-2,5-dione in this context. δ-Carbolines fused with pyrrolidine-2,5-dione are of interest because the succinimide part of fused heteroaromatic molecule can interact with the ATP binding pocket via the hydrogen bond network with selectivity towards α-Topo II. The 300 derivatives designed were subjected to the Lipinski rule of 5, ADMET and toxicity prediction. The designed compounds were further analysed using molecular docking analysis on the active sites of the α-Topo II crystal structure (PDB ID:1ZXM). Molecular dynamic simulations were also performed to compare the binding mode and stability of the protein-ligand complexes. Compounds with ID numbers AS89, AS104, AS119, AS209, AS239, AS269, and AS299 show good binding activity compared to the co-crystal ligand. Molecular Dynamics simulation studies show that the ligand binding to α-Topo II in the ATP domain is stableand the protein-ligand conformation remains unchanged. Binding free energy calculations suggest that seven molecules designed are potential inhibitors for α-Topo II for further consideration as anticancer agents.
Collapse
Affiliation(s)
- Selvaraj Ayyamperumal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris - 643001,Tamil Nadu, India
| | - Dhananjay DJ
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi - 110067, India
| | - Vyshnavi Tallapaneni
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris - 643001,Tamil Nadu, India
| | - Surender Mohan
- Laboratory of Molecular Biology and Genetic Engineering, School of Biotechnology, Jawaharlal Nehru University, New Delhi - 110067, India
| | - Basappa S
- Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore - 570006, Karnataka, India
| | - Jubie Selvaraj
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris - 643001,Tamil Nadu, India
| | - Nanjan Moola Joghee
- PG Studies and Research, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris - 643001, Tamil Nadu, India
| | - Chandrasekar MJN
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris - 643001,Tamil Nadu, India
| |
Collapse
|
21
|
Buzun K, Bielawska A, Bielawski K, Gornowicz A. DNA topoisomerases as molecular targets for anticancer drugs. J Enzyme Inhib Med Chem 2020; 35:1781-1799. [PMID: 32975138 PMCID: PMC7534307 DOI: 10.1080/14756366.2020.1821676] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
The significant role of topoisomerases in the control of DNA chain topology has been confirmed in numerous research conducted worldwide. The prevalence of these enzymes, as well as the key importance of topoisomerase in the proper functioning of cells, have made them the target of many scientific studies conducted all over the world. This article is a comprehensive review of knowledge about topoisomerases and their inhibitors collected over the years. Studies on the structure-activity relationship and molecular docking are one of the key elements driving drug development. In addition to information on molecular targets, this article contains details on the structure-activity relationship of described classes of compounds. Moreover, the work also includes details about the structure of the compounds that drive the mode of action of topoisomerase inhibitors. Finally, selected topoisomerases inhibitors at the stage of clinical trials and their potential application in the chemotherapy of various cancers are described.
Collapse
Affiliation(s)
- Kamila Buzun
- Department of Biotechnology, Medical University of Bialystok, Bialystok, Poland
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Bialystok, Bialystok, Poland
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Bialystok, Poland
| | - Agnieszka Gornowicz
- Department of Biotechnology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
22
|
Synthesis, characterization, and hypoglycemic efficacy of nitro and amino acridines and 4-phenylquinoline on starch hydrolyzing compounds: an in silico and in vitro study. Struct Chem 2020. [DOI: 10.1007/s11224-020-01529-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
23
|
Kozurkova M, Sabolova D, Kristian P. A new look at 9-substituted acridines with various biological activities. J Appl Toxicol 2020; 41:175-189. [PMID: 32969520 DOI: 10.1002/jat.4072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/28/2022]
Abstract
Heterocycles have long been the focus of intensive study in attempts to develop novel therapeutic compounds, and acridine, a polynuclear nitrogen molecule containing a heterocycle, has attracted a considerable amount of scientific attention. Acridine derivatives have been studied in detail and have been found to possess multitarget properties, which inhibit topoisomerase enzymes that regulate topological changes in DNA and interfere with the essential biological function of DNA. This article describes some recent advancements in the field of new 9-substituted acridine heterocyclic agents and describes both the structure and the structure-activity relationship of the most promising molecules. The article will also present the IC50 values of the novel derivatives against various human cancer cell lines. The mini review also investigates the topoisomerase inhibition and antibacterial and antimalarial activity of these polycyclic aromatic derivatives.
Collapse
Affiliation(s)
- Maria Kozurkova
- Department of Biochemistry, Institute of Chemistry, Faculty of Science, P. J. Šafárik University, Kosice, Slovak Republic.,Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Danica Sabolova
- Department of Biochemistry, Institute of Chemistry, Faculty of Science, P. J. Šafárik University, Kosice, Slovak Republic
| | - Pavol Kristian
- Department of Organic Chemistry, Institute of Chemistry, Faculty of Science, P. J. Šafárik University, Kosice, Slovak Republic
| |
Collapse
|
24
|
Bailly C. Pyronaridine: An update of its pharmacological activities and mechanisms of action. Biopolymers 2020; 112:e23398. [PMID: 33280083 DOI: 10.1002/bip.23398] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
Pyronaridine (PYR) is an erythrocytic schizonticide with a potent antimalarial activity against multidrug-resistant Plasmodium. The drug is used in combination with artesunate for the treatment of uncomplicated P. falciparum malaria, in adults and children. The present review briefly retraces the discovery of PYR and recent antimalarial studies which has led to the approval of PYR/artesunate combination (Pyramax) by the European Medicines Agency to treat uncomplicated malaria worldwide. PYR also presents a marked antitumor activity and has revealed efficacy for the treatment of other parasitic diseases (notably Babesia and Trypanosoma infections) and to mitigate the Ebola virus propagation. On the one hand, PYR functions has an inhibitor of hemozoin (biomineral malaria pigment, by-product of hemoglobin digestion) formation, blocking the biopolymerization of β-hematin and thus facilitating the accumulation of toxic hematin into the digestive vacuole of the parasite. On the other hand, PYR is a bona fide DNA-intercalating agent and an inhibitor of DNA topoisomerase 2, leading to DNA damages and cell death. Inhibition of hematin polymerization represents the prime mechanism at the origin of the antimalarial activity, whereas anticancer effects relies essentially on the interference with DNA metabolism, as with structurally related anticancer drugs like amsacrine and quinacrine. In addition, recent studies point to an immune modulatory activity of PYR and the implication of a mitochondrial oxidative pathway. An analogy with the mechanism of action of artemisinin drugs is underlined. In brief, the biological actions of pyronaridine are recapitulated to shed light on the diverse health benefits of this unsung drug.
Collapse
|
25
|
Cichorek M, Ronowska A, Dzierzbicka K, Gensicka-Kowalewska M, Deptula M, Pelikant-Malecka I. Chloroacridine derivatives as potential anticancer agents which may act as tricarboxylic acid cycle enzyme inhibitors. Biomed Pharmacother 2020; 130:110515. [PMID: 34321163 DOI: 10.1016/j.biopha.2020.110515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 10/23/2022] Open
Abstract
PURPOSE This paper concerns the cytotoxicity of 9-chloro-1-nitroacridine (1a) and 9-chloro-4-methyl-1-nitroacridine (1b) against two biologically different melanoma forms: melanotic and amelanotic. Melanomas are tumors characterized by high heterogeneity and poor susceptibility to chemotherapies. Among new analogs synthesized by us, compound 1b exhibited the highest anticancer potency. Because of that, in this study, we analyzed the mechanism of action for 1a and its 4-methylated derivative, 1b, against a pair of biological melanoma forms, with regard to proliferation, cell death mechanism and energetic state. METHODS Cytotoxicity was evaluated by XTT assay. Cell death was estimated by plasma membrane structure changes (phosphatidylserine externalization), caspase activation, and ROS presence. The energetic state of cells was estimated based on NAD and ATP levels, and the activity of tricarboxylic acid cycle enzymes (pyruvate dehydrogenase complex, aconitase, isocitrate dehydrogenase). RESULTS The chloroacridines affect biological forms of melanoma in different ways. Amelanotic (Ab) melanoma (with inhibited melanogenesis and higher malignancy) was particularly sensitive to the action of the chloroacridines. The Ab melanoma cells died through apoptosis and through death without caspase activation. Diminished activity of TAC enzymes was noticed among Ab melanoma cells together with ATP/NAD depletion, especially in the case of 1b. CONCLUSION Our data show that the biological forms of the tumors responded to 1a and its 4-methylated analog in different ways. 1a and 1b could be inducers of regulated melanoma cell death, especially the amelanotic form. Although the mechanism of the cell death is not fully understood, 1b may act by interfering with the TAC enzymes and blocking specific pathways leading to tumor growth. This could encourage further investigation of its anticancer activity, especially against the amelanotic form of melanoma.
Collapse
Affiliation(s)
- Miroslawa Cichorek
- Department of Embryology, Medical University of Gdansk, Debinki 1 St. PL, 80-210, Gdansk, Poland.
| | - Anna Ronowska
- Department of Laboratory Medicine, Medical University of Gdansk, Debinki 7 St. PL, 80-211, Gdansk, Poland
| | - Krystyna Dzierzbicka
- Department of Organic Chemistry, Gdansk University of Technology, Narutowicza St. 11/12. PL, 80-233, Gdansk, Poland
| | - Monika Gensicka-Kowalewska
- Department of Organic Chemistry, Gdansk University of Technology, Narutowicza St. 11/12. PL, 80-233, Gdansk, Poland
| | - Milena Deptula
- Department of Embryology, Medical University of Gdansk, Debinki 1 St. PL, 80-210, Gdansk, Poland
| | - Iwona Pelikant-Malecka
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 St. PL, 80-210, Gdansk, Poland; Department of Medical Laboratory Diagnostics, Central Bank of Frozen Tissues and Genetic Specimens, Medical University of Gdansk, Biobanking and Biomolecular Resources Research Infrastructure Poland, Debinki 7 St. PL, 80-211, Gdansk, Poland
| |
Collapse
|
26
|
The Gene scb-1 Underlies Variation in Caenorhabditis elegans Chemotherapeutic Responses. G3-GENES GENOMES GENETICS 2020; 10:2353-2364. [PMID: 32385045 PMCID: PMC7341127 DOI: 10.1534/g3.120.401310] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pleiotropy, the concept that a single gene controls multiple distinct traits, is prevalent in most organisms and has broad implications for medicine and agriculture. The identification of the molecular mechanisms underlying pleiotropy has the power to reveal previously unknown biological connections between seemingly unrelated traits. Additionally, the discovery of pleiotropic genes increases our understanding of both genetic and phenotypic complexity by characterizing novel gene functions. Quantitative trait locus (QTL) mapping has been used to identify several pleiotropic regions in many organisms. However, gene knockout studies are needed to eliminate the possibility of tightly linked, non-pleiotropic loci. Here, we use a panel of 296 recombinant inbred advanced intercross lines of Caenorhabditis elegans and a high-throughput fitness assay to identify a single large-effect QTL on the center of chromosome V associated with variation in responses to eight chemotherapeutics. We validate this QTL with near-isogenic lines and pair genome-wide gene expression data with drug response traits to perform mediation analysis, leading to the identification of a pleiotropic candidate gene, scb-1, for some of the eight chemotherapeutics. Using deletion strains created by genome editing, we show that scb-1, which was previously implicated in response to bleomycin, also underlies responses to other double-strand DNA break-inducing chemotherapeutics. This finding provides new evidence for the role of scb-1 in the nematode drug response and highlights the power of mediation analysis to identify causal genes.
Collapse
|
27
|
Rupar J, Dobričić V, Grahovac J, Radulović S, Skok Ž, Ilaš J, Aleksić M, Brborić J, Čudina O. Synthesis and evaluation of anticancer activity of new 9-acridinyl amino acid derivatives. RSC Med Chem 2020; 11:378-386. [PMID: 33479643 DOI: 10.1039/c9md00597h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 01/19/2020] [Indexed: 01/25/2023] Open
Abstract
A series of eleven 9-acridinyl amino acid derivatives were synthesized using a two-step procedure. Cytotoxicity was tested on the K562 and A549 cancer cell lines and normal diploid cell line MRC5 using the MTT assay. Compounds 6, 7, 8 and 9 were the most active, with IC50 values comparable to or lower than that of chemotherapeutic agent amsacrine. 8 and 9 were especially effective in the A549 cell line (IC50 ≈ 6 μM), which is of special interest since amsacrine is not sufficiently active in lung cancer patients. Cell cycle analysis revealed that 7 and 9 caused G2/M block, amsacrine caused arrest in the S phase, while 6 and 8 induced apoptotic cell death independently of the cell cycle regulation. In comparison to amsacrine, 6, 7, 8, and 9 showed similar inhibitory potential towards topoisomerase II, whereas only 7 showed DNA intercalation properties. In contrast to amsacrine, 6, 7, 8 and 9 showed a lack of toxicity towards unstimulated normal human leucocytes.
Collapse
Affiliation(s)
- Jelena Rupar
- Department of Pharmaceutical Chemistry , University of Belgrade - Faculty of Pharmacy , Vojvode Stepe 450 , 11000 Belgrade , Serbia . .,Department of Physical Chemistry and Instrumental Methods , University of Belgrade - Faculty of Pharmacy , Vojvode Stepe 450 , 11000 Belgrade , Serbia
| | - Vladimir Dobričić
- Department of Pharmaceutical Chemistry , University of Belgrade - Faculty of Pharmacy , Vojvode Stepe 450 , 11000 Belgrade , Serbia .
| | - Jelena Grahovac
- Department of Experimental Oncology , Institute for Oncology and Radiology of Serbia , Pasterova 14 , Belgrade , Serbia
| | - Siniša Radulović
- Department of Experimental Oncology , Institute for Oncology and Radiology of Serbia , Pasterova 14 , Belgrade , Serbia
| | - Žiga Skok
- Chair of Pharmaceutical Chemistry , University of Ljubljana, Faculty of Pharmacy , Aškerčeva 7 , SI-1000 Ljubljana , Slovenia
| | - Janez Ilaš
- Chair of Pharmaceutical Chemistry , University of Ljubljana, Faculty of Pharmacy , Aškerčeva 7 , SI-1000 Ljubljana , Slovenia
| | - Mara Aleksić
- Department of Physical Chemistry and Instrumental Methods , University of Belgrade - Faculty of Pharmacy , Vojvode Stepe 450 , 11000 Belgrade , Serbia
| | - Jasmina Brborić
- Department of Pharmaceutical Chemistry , University of Belgrade - Faculty of Pharmacy , Vojvode Stepe 450 , 11000 Belgrade , Serbia .
| | - Olivera Čudina
- Department of Pharmaceutical Chemistry , University of Belgrade - Faculty of Pharmacy , Vojvode Stepe 450 , 11000 Belgrade , Serbia .
| |
Collapse
|
28
|
Sivaramakarthikeyan R, Iniyaval S, Lim WM, Hii LW, Mai CW, Ramalingan C. Pyrazolylphenanthroimidazole heterocycles: synthesis, biological and molecular docking studies. NEW J CHEM 2020; 44:19612-19622. [DOI: 10.1039/d0nj02214d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2025]
Abstract
The synthesis of a series of novel pyrazolylphenanthroimidazoles 6a–6j has been accomplished utilizing a multi-step synthetic protocol, and characterized through physical and spectral techniques.
Collapse
Affiliation(s)
- Ramar Sivaramakarthikeyan
- Department of Chemistry
- School of Advanced Sciences
- Kalasalingam Academy of Research and Education (Deemed to be University)
- Krishnankoil
- India
| | - Shunmugam Iniyaval
- Department of Chemistry
- School of Advanced Sciences
- Kalasalingam Academy of Research and Education (Deemed to be University)
- Krishnankoil
- India
| | - Wei-Meng Lim
- School of Pharmacy
- International Medical University
- Bukit Jalil
- Malaysia
| | - Ling-Wei Hii
- School of Pharmacy
- International Medical University
- Bukit Jalil
- Malaysia
| | - Chun-Wai Mai
- School of Pharmacy
- International Medical University
- Bukit Jalil
- Malaysia
- Center for Cancer and Stem Cell Research
| | - Chennan Ramalingan
- Department of Chemistry
- School of Advanced Sciences
- Kalasalingam Academy of Research and Education (Deemed to be University)
- Krishnankoil
- India
| |
Collapse
|
29
|
Ribeiro AG, Almeida SMVD, de Oliveira JF, Souza TRCDL, Santos KLD, Albuquerque APDB, Nogueira MCDBL, Carvalho Junior LBD, Moura ROD, da Silva AC, Pereira VRA, Castro MCABD, Lima MDCAD. Novel 4-quinoline-thiosemicarbazone derivatives: Synthesis, antiproliferative activity, in vitro and in silico biomacromolecule interaction studies and topoisomerase inhibition. Eur J Med Chem 2019; 182:111592. [DOI: 10.1016/j.ejmech.2019.111592] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/07/2019] [Accepted: 08/05/2019] [Indexed: 12/16/2022]
|
30
|
Ismail NA, Salman AA, Mohd Yusof MS, Che Soh SK, Kadir Pahirulzaman KA, Ali HM, Sarip R. Synthesis, cytotoxicity and antineoplastic activities of novel acridine-based platinum(II) organometallic complexes. J Organomet Chem 2019. [DOI: 10.1016/j.jorganchem.2019.06.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
31
|
Gouveia RG, Ribeiro AG, Segundo MÂSP, de Oliveira JF, de Lima MDCA, de Lima Souza TRC, de Almeida SMV, de Moura RO. Synthesis, DNA and protein interactions and human topoisomerase inhibition of novel Spiroacridine derivatives. Bioorg Med Chem 2018; 26:5911-5921. [DOI: 10.1016/j.bmc.2018.10.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/24/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022]
|
32
|
Hevener K, Verstak TA, Lutat KE, Riggsbee DL, Mooney JW. Recent developments in topoisomerase-targeted cancer chemotherapy. Acta Pharm Sin B 2018; 8:844-861. [PMID: 30505655 PMCID: PMC6251812 DOI: 10.1016/j.apsb.2018.07.008] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 12/17/2022] Open
Abstract
The DNA topoisomerase enzymes are essential to cell function and are found ubiquitously in all domains of life. The various topoisomerase enzymes perform a wide range of functions related to the maintenance of DNA topology during DNA replication, and transcription are the targets of a wide range of antimicrobial and cancer chemotherapeutic agents. Natural product-derived agents, such as the camptothecin, anthracycline, and podophyllotoxin drugs, have seen broad use in the treatment of many types of cancer. Selective targeting of the topoisomerase enzymes for cancer treatment continues to be a highly active area of basic and clinical research. The focus of this review will be to summarize the current state of the art with respect to clinically used topoisomerase inhibitors for targeted cancer treatment and to discuss the pharmacology and chemistry of promising new topoisomerase inhibitors in clinical and pre-clinical development.
Collapse
Affiliation(s)
- KirkE. Hevener
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | |
Collapse
|
33
|
Szafran MJ, Kołodziej M, Skut P, Medapi B, Domagała A, Trojanowski D, Zakrzewska-Czerwińska J, Sriram D, Jakimowicz D. Amsacrine Derivatives Selectively Inhibit Mycobacterial Topoisomerase I (TopA), Impair M. smegmatis Growth and Disturb Chromosome Replication. Front Microbiol 2018; 9:1592. [PMID: 30065714 PMCID: PMC6056748 DOI: 10.3389/fmicb.2018.01592] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/26/2018] [Indexed: 01/21/2023] Open
Abstract
Amsacrine, which inhibits eukaryotic type II topoisomerase via DNA intercalation and stabilization of the cleavable topoisomerase-DNA complex, promotes DNA damage and eventually cell death. Amsacrine has also been shown to inhibit structurally distinct bacterial type I topoisomerases (TopAs), including mycobacterial TopA, the only and essential topoisomerase I in Mycobacterium tuberculosis. Here, we describe the modifications of an amsacrine sulfonamide moiety that presumably interacts with mycobacterial TopA, which notably increased the enzyme inhibition and drug selectivity in vivo. To analyse the effects of amsacrine and its derivatives treatment on cell cycle, we used time-lapse fluorescence microscopy (TLMM) and fusion of the β-subunit of DNA polymerase III with enhanced green fluorescence protein (DnaN-EGFP). We determined that treatment with amsacrine and its derivatives increased the number of DnaN-EGFP complexes and/or prolonged the time of chromosome replication and cell cycle notably. The analysis of TopA depletion strain confirmed that lowering TopA level results in similar disturbances of chromosome replication. In summary, since TopA is crucial for mycobacterial cell viability, the compounds targeting the enzyme disturbed the cell cycle and thus may constitute a new class of anti-tuberculosis drugs.
Collapse
Affiliation(s)
- Marcin J Szafran
- Laboratory of Molecular Microbiology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Marta Kołodziej
- Laboratory of Molecular Microbiology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Patrycja Skut
- Laboratory of Molecular Microbiology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Brahmam Medapi
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | | | - Damian Trojanowski
- Laboratory of Molecular Microbiology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Jolanta Zakrzewska-Czerwińska
- Laboratory of Molecular Microbiology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland.,Laboratory of Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Dharmarajan Sriram
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad, India
| | - Dagmara Jakimowicz
- Laboratory of Molecular Microbiology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland.,Laboratory of Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| |
Collapse
|
34
|
N-(acridin-9-yl)arenesulfonamides: Synthesis, quantum chemical studies and crystal structure analysis to establish the tautomeric preferences. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.05.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
35
|
Zhou Q, You C, Zheng C, Gu Y, Gu H, Zhang R, Wu H, Sun B. 3-Nitroacridine derivatives arrest cell cycle at G0/G1 phase and induce apoptosis in human breast cancer cells may act as DNA-target anticancer agents. Life Sci 2018; 206:1-9. [PMID: 29738780 DOI: 10.1016/j.lfs.2018.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/03/2018] [Accepted: 05/03/2018] [Indexed: 12/14/2022]
Abstract
DNA is considered to be one of the most promising targets for anticancer agents. Acridine analogues have anticancer activity based on DNA binding and topoisomerases inhibition. However, due to the side effects, resistance and low bioavailability, a few have entered into clinical usage and the mechanisms of action are not fully understood. Novel acridine derivatives are needed for effective cancer therapy. A series of novel 3-nitroacridine-based derivatives were synthesized, their DNA binding and anticancer activities were evaluated. The chemical modifications at position 9 of the 3-nitroacridine were crucial for DNA affinity, thus optimizing anticancer activity. UV-Vis and circular dichroism (CD) spectroscopy indicated interaction of compounds with DNA, and the binding modes were intercalation and groove binding. MTT assay and clonogenic assay showed that compounds 1, 2 and 3 had obvious cell growth inhibition effect. They induced cell apoptosis in human breast cancer cells in a dose-dependent manner, and exhibited anticancer effect via DNA damage as well as cell cycle arrest at G0/G1 phage. Using confocal fluorescent microscope, the apoptotic features were observed. The results suggested that compounds 1-3 with high DNA binding affinity and good inhibitory effect of cancer cell proliferation can be developed as prime candidates for further chemical optimization.
Collapse
Affiliation(s)
- Qian Zhou
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, China
| | - Chaoqun You
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, China
| | - Cong Zheng
- Department of Chemical and Pharmaceutical Engineering, Southeast University Chenxian College, Nanjing 210000, China
| | - Yawen Gu
- Department of Chemical and Pharmaceutical Engineering, Southeast University Chenxian College, Nanjing 210000, China
| | - Hongchao Gu
- Department of Chemical and Pharmaceutical Engineering, Southeast University Chenxian College, Nanjing 210000, China
| | - Rui Zhang
- Department of Chemical and Pharmaceutical Engineering, Southeast University Chenxian College, Nanjing 210000, China
| | - Hongshuai Wu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, China
| | - Baiwang Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, China.
| |
Collapse
|
36
|
Sonawane AD, Garud DR, Udagawa T, Kubota Y, Koketsu M. Synthesis of thieno[2,3-c]acridine and furo[2,3-c]acridine derivatives via an iodocyclization reaction and their fluorescence properties and DFT mechanistic studies. NEW J CHEM 2018. [DOI: 10.1039/c8nj03511c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In this paper, we report the novel synthesis of thieno[2,3-c]acridine and furo[2,3-c]acridine derivatives via intramolecular iodocyclization reaction. The thieno[2,3-c]acridine derivatives exhibited blue fluorescence in hexane.
Collapse
Affiliation(s)
- Amol D. Sonawane
- Department of Chemistry and Biomolecular Science
- Faculty of Engineering
- Gifu University
- Gifu 501-1193
- Japan
| | - Dinesh R. Garud
- Department of Chemistry
- Sir Parashurambhau College
- Pune 411030
- India
| | - Taro Udagawa
- Department of Chemistry and Biomolecular Science
- Faculty of Engineering
- Gifu University
- Gifu 501-1193
- Japan
| | - Yasuhiro Kubota
- Department of Chemistry and Biomolecular Science
- Faculty of Engineering
- Gifu University
- Gifu 501-1193
- Japan
| | - Mamoru Koketsu
- Department of Chemistry and Biomolecular Science
- Faculty of Engineering
- Gifu University
- Gifu 501-1193
- Japan
| |
Collapse
|
37
|
Harada K, Imai T, Kizu J, Mochizuki M, Inami K. DNA interaction of bromomethyl-substituted acridines. Med Chem Res 2017. [DOI: 10.1007/s00044-017-2030-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
38
|
de Almeida SMV, Ribeiro AG, de Lima Silva GC, Ferreira Alves JE, Beltrão EIC, de Oliveira JF, de Carvalho LB, Alves de Lima MDC. DNA binding and Topoisomerase inhibition: How can these mechanisms be explored to design more specific anticancer agents? Biomed Pharmacother 2017; 96:1538-1556. [DOI: 10.1016/j.biopha.2017.11.054] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 12/11/2022] Open
|
39
|
Khadka DB, Park S, Jin Y, Han J, Kwon Y, Cho WJ. Design, synthesis, and biological evaluation of 1,3-diarylisoquinolines as novel topoisomerase I catalytic inhibitors. Eur J Med Chem 2017; 143:200-215. [PMID: 29174815 DOI: 10.1016/j.ejmech.2017.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/03/2017] [Accepted: 11/04/2017] [Indexed: 12/30/2022]
Abstract
With a goal of identifying potent topoisomerase (topo) inhibitor, the C4-aromatic ring of the anticancer agent, 3,4-diarylisoquinolone, was strategically shifted to design 1,3-diarylisoquinoline. Twenty-two target compounds were synthesized in three simple and efficient steps. The 1,3-diarylisoquinolines exhibited potent anti-proliferative effects on cancer cells but few compounds spared non-cancerous cells. Inhibition of topo I/IIα-mediated DNA relaxation by several derivatives was greater than that by camptothecin (CPT)/etoposide even at low concentration (20 μM). In addition, these compounds had little or no effect on polymerization of tubulin. A series of biological evaluations performed with the most potent derivative 4cc revealed that the compound is a non-intercalative topo I catalytic inhibitor interacting with free topo I. Collectively, the potent cytotoxic effect on cancer cells including the drug resistance ones, absence of lethal effect on normal cells, and different mechanism of action than topo I poisons suggest that the 1,3-diarylisoquinolines might be a promising class of anticancer agents worthy of further pursuit.
Collapse
Affiliation(s)
- Daulat Bikram Khadka
- College of Pharmacy, Research Institute of Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seojeong Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yifeng Jin
- College of Pharmacy, Research Institute of Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jinhe Han
- College of Pharmacy, Research Institute of Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Youngjoo Kwon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Won-Jea Cho
- College of Pharmacy, Research Institute of Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
40
|
Ali I, Lone MN, Aboul-Enein HY. Imidazoles as potential anticancer agents. MEDCHEMCOMM 2017; 8:1742-1773. [PMID: 30108886 PMCID: PMC6084102 DOI: 10.1039/c7md00067g] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/21/2017] [Indexed: 12/19/2022]
Abstract
Cancer is a black spot on the face of humanity in this era of science and technology. Presently, several classes of anticancer drugs are available in the market, but issues such as toxicity, low efficacy and solubility have decreased the overall therapeutic indices. Thus, the search for new promising anticancer agents continues, and the battle against cancer is far from over. Imidazole is an aromatic diazole and alkaloid with anticancer properties. There is considerable interest among scientists in developing imidazoles as safe alternatives to anticancer chemotherapy. The present article describes the structural, chemical, and biological features of imidazoles. Several classes of imidazoles as anticancer agents based on their mode of action have been critically discussed. A careful observation has been made into pharmacologically active imidazoles with better or equal therapeutic effects compared to well-known imidazole-based anticancer drugs, which are available on the market. A brief discussion of the toxicities of imidazoles has been made. Finally, the current challenges and future perspectives of imidazole based anticancer drug development are conferred.
Collapse
Affiliation(s)
- Imran Ali
- Department of Chemistry , Jamia Millia Islamia (Central University) , New Delhi-110025 , India . ;
| | - Mohammad Nadeem Lone
- Department of Chemistry , Jamia Millia Islamia (Central University) , New Delhi-110025 , India . ;
| | - Haasan Y Aboul-Enein
- Pharmaceutical and Medicinal Chemistry Department , Pharmaceutical and Drug Industries Research Division , National Research Centre , Dokki , Giza 12622 , Egypt
| |
Collapse
|
41
|
Ranjan N, Story S, Fulcrand G, Leng F, Ahmad M, King A, Sur S, Wang W, Tse-Dinh YC, Arya DP. Selective Inhibition of Escherichia coli RNA and DNA Topoisomerase I by Hoechst 33258 Derived Mono- and Bisbenzimidazoles. J Med Chem 2017; 60:4904-4922. [DOI: 10.1021/acs.jmedchem.7b00191] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Nihar Ranjan
- Laboratory
of Medicinal Chemistry, Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Sandra Story
- NUBAD LLC, 900B West Faris
Road, Greenville, South Carolina 29605, United States
| | - Geraldine Fulcrand
- Department
of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
- Biomolecular
Sciences Institute, Florida International University, Miami, Florida 33199, United States
| | - Fenfei Leng
- Department
of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
- Biomolecular
Sciences Institute, Florida International University, Miami, Florida 33199, United States
| | - Muzammil Ahmad
- Genome
Instability and Chromatin Remodeling Section, Lab of Genetics, National
Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, Maryland 21224, United States
| | - Ada King
- NUBAD LLC, 900B West Faris
Road, Greenville, South Carolina 29605, United States
| | - Souvik Sur
- Laboratory
of Medicinal Chemistry, Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Weidong Wang
- Genome
Instability and Chromatin Remodeling Section, Lab of Genetics, National
Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, Maryland 21224, United States
| | - Yuk-Ching Tse-Dinh
- Department
of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
- Biomolecular
Sciences Institute, Florida International University, Miami, Florida 33199, United States
| | - Dev P. Arya
- Laboratory
of Medicinal Chemistry, Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
- NUBAD LLC, 900B West Faris
Road, Greenville, South Carolina 29605, United States
| |
Collapse
|
42
|
Musiol R. An overview of quinoline as a privileged scaffold in cancer drug discovery. Expert Opin Drug Discov 2017; 12:583-597. [DOI: 10.1080/17460441.2017.1319357] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
43
|
Mangueira VM, Batista TM, Brito MT, Sousa TKGD, Cruz RMDD, Abrantes RAD, Veras RC, Medeiros IAD, Medeiros KKDP, Pereira ALDC, Serafim VDL, Moura ROD, Sobral MV. A new acridine derivative induces cell cycle arrest and antiangiogenic effect on Ehrlich ascites carcinoma model. Biomed Pharmacother 2017; 90:253-261. [PMID: 28364597 DOI: 10.1016/j.biopha.2017.03.049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 03/07/2017] [Accepted: 03/18/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Acridine derivatives, including amsacrine, have antitumor activity. However, side effects, development of resistance and their low bioavailability, have limited their use. Herein, we described the synthesis, and evaluated the toxicity and antitumor activity of a new amsacrine analogous, the N'-(2-chloro-6-methoxy-acridin-9-yl)-2-cyano-3-(4-dimethylaminophenyl)-acrilohidrazida (ACS-AZ10). METHODS The compound was obtained in a linear pathway where the ASC-Az intermediate was obtained by coupling of 6,9-dichloro-3-methoxy-acridine and 2-ciany-acethohidrazide followed by condensation with the corresponding aldehyde. The toxicity of ACS-AZ10 was evaluated in mice using acute toxicity and micronucleus assays. Ehrlich ascites carcinoma model was used to investigate the antitumor activity and toxicity of ACS-AZ10 (7.5, 15 or 30mg/kg, i.p.), after nine days of treatment. Cell cycle and angiogenesis were also evaluated. RESULTS The ASC-AZ10 was obtained with satisfactory yields and its structure was confirmed by spectroscopic and spectrometric techniques. On acute toxicity study, ACS-AZ10 (2000mg/kg, i.p.) induced transient depressant effects on central nervous system. The LD50 was approximately 2500mg/kg. ACS-AZ10 (15 or 30mg/kg) displayed significant antitumor activity considering the tumor weight and volume, cell viability, and total Ehrlich cell count. ACS-AZ10 (7.5mg/kg) induced an increase in sub-G1 peak, suggesting apoptosis. At 15mg/kg ACS-AZ10 induced cell cycle arrest in G2/M phase and a reduction in the percentage of cells in G0/G1 and S phases, suggesting a pre-mitotic blockade. ACS-AZ10 reduced the microvessel density, indicating an antiangiogenic effect. Weak hematological, biochemical and histopathological toxicity were observed. The compound doesn't show genotoxicity in micronucleus assay. CONCLUSIONS ACS-AZ10 has potent antitumor activity in vivo along with low toxicity.
Collapse
Affiliation(s)
- Vivianne Mendes Mangueira
- Programa de Pós Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil, Brazil.
| | - Tatianne Mota Batista
- Programa de Pós Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil, Brazil.
| | - Monalisa Taveira Brito
- Programa de Pós Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil, Brazil.
| | - Tatyanna Kelvia Gomes de Sousa
- Programa de Pós Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil, Brazil.
| | - Ryldene Marques Duarte da Cruz
- Programa de Pós Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil, Brazil.
| | - Renata Albuquerque de Abrantes
- Programa de Pós Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil, Brazil.
| | - Robson Cavalcanti Veras
- Programa de Pós Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil, Brazil; Departamento de Ciências Farmacêuticas, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil.
| | - Isac Almeida de Medeiros
- Programa de Pós Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil, Brazil; Departamento de Ciências Farmacêuticas, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil.
| | | | - Ana Ligia da Costa Pereira
- Laboratório de Sintese e Vetorização Molecular, Departamento de Farmácia, Universidade Estadual da Paraíba, 58429-500 Campina Grande, Paraíba, Brazil.
| | - Vanessa de Lima Serafim
- Laboratório de Sintese e Vetorização Molecular, Departamento de Farmácia, Universidade Estadual da Paraíba, 58429-500 Campina Grande, Paraíba, Brazil.
| | - Ricardo Olímpio de Moura
- Laboratório de Sintese e Vetorização Molecular, Departamento de Farmácia, Universidade Estadual da Paraíba, 58429-500 Campina Grande, Paraíba, Brazil.
| | - Marianna Vieira Sobral
- Programa de Pós Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil, Brazil; Departamento de Ciências Farmacêuticas, Universidade Federal da Paraíba, 58051-970 João Pessoa, Paraíba, Brazil.
| |
Collapse
|
44
|
Sader S, Wu C. Computational analysis of Amsacrine resistance in human topoisomerase II alpha mutants (R487K and E571K) using homology modeling, docking and all-atom molecular dynamics simulation in explicit solvent. J Mol Graph Model 2017; 72:209-219. [DOI: 10.1016/j.jmgm.2016.11.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/07/2016] [Accepted: 11/18/2016] [Indexed: 11/30/2022]
|
45
|
Cestrum nocturnum Flower Extracts Attenuate Proliferation and Induce Apoptosis in Malignant Cells through Inducing DNA Damage and Inhibiting Topoisomerase II Activity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:1456786. [PMID: 28250789 PMCID: PMC5307125 DOI: 10.1155/2017/1456786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 12/20/2016] [Indexed: 11/18/2022]
Abstract
Most of the existing chemotherapeutic drugs have plenty of side effects. Chinese herbal medicine has been used for pharmaceutical and dietary therapy for thousands of years with more effective and fewer side effects. Cestrum nocturnum (CN) has long been used to treat digestive diseases for centuries in China. Our previous study first proved that the n-butanol part isolated from the flowers of CN produced an inhibitory effect on the proliferation of malignant cells. However, the fractions responsible for the antiproliferation effect of n-butanol part from CN flowers and related mechanisms remain unknown. Thus, in this study, we extracted fractions C4 and C5 from n-butanol part of CN flowers and investigated their immune toxicity and antitumor activities. It was found that fractions C4 and C5 exhibited great cytotoxicity to cancer cell lines but had low immune toxicity towards T and B lymphocytes in vitro. The tested fractions also attenuated proliferation and induced apoptosis at G0/G1 and G2/M phases in Bel-7404 cells through inducing DNA damage and inhibiting topoisomerase II relaxation activity. These results suggest that fractions C4 and C5 may represent important sources of potential antitumor agents due to their pronounced antitumor effects and low immune toxicity.
Collapse
|
46
|
|
47
|
Infante Lara L, Sledge A, Laradji A, Okoro CO, Osheroff N. Novel trifluoromethylated 9-amino-3,4-dihydroacridin-1(2H)-ones act as covalent poisons of human topoisomerase IIα. Bioorg Med Chem Lett 2016; 27:586-589. [PMID: 27998679 DOI: 10.1016/j.bmcl.2016.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/02/2016] [Accepted: 12/02/2016] [Indexed: 10/20/2022]
Abstract
A number of topoisomerase II-targeted anticancer drugs, including amsacrine, utilize an acridine or related aromatic core as a scaffold. Therefore, to further explore the potential of acridine-related compounds to act as topoisomerase II poisons, we synthesized a series of novel trifluoromethylated 9-amino-3,4-dihydroacridin-1(2H)-one derivatives and examined their ability to enhance DNA cleavage mediated by human topoisomerase IIα. Derivatives containing a H, Cl, F, and Br at C7 enhanced enzyme-mediated double-stranded DNA cleavage ∼5.5- to 8.5-fold over baseline, but were less potent than amsacrine. The inclusion of an amino group at C9 was critical for activity. The compounds lost their activity against topoisomerase IIα in the presence of a reducing agent, displayed no activity against the catalytic core of topoisomerase IIα, and inhibited DNA cleavage when incubated with the enzyme prior to the addition of DNA. These findings strongly suggest that the compounds act as covalent, rather than interfacial, topoisomerase II poisons.
Collapse
Affiliation(s)
- Lorena Infante Lara
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Alexis Sledge
- Department of Chemistry, Tennessee State University, Nashville, TN 37209-1561, USA
| | - Amine Laradji
- Department of Chemistry, Tennessee State University, Nashville, TN 37209-1561, USA
| | - Cosmas O Okoro
- Department of Chemistry, Tennessee State University, Nashville, TN 37209-1561, USA.
| | - Neil Osheroff
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Medicine (Hematology/Oncology), Vanderbilt University School of Medicine, Nashville, TN 37232, USA; VA Tennessee Valley Healthcare System, Nashville, TN 37212, USA.
| |
Collapse
|
48
|
Chagas MBO, Cordeiro NCC, Marques KMR, Rocha Pitta MG, Rêgo MJBM, Lima MCA, Pitta MGR, Pitta IR. New thiazacridine agents: Synthesis, physical and chemical characterization, and in vitro anticancer evaluation. Hum Exp Toxicol 2016; 36:1059-1070. [DOI: 10.1177/0960327116680274] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A series of new thiazacridine agents were synthesized and evaluated as antitumor agents, in terms of not only their cytotoxicity but also their selectivity. The cytotoxicity assay confirmed that all compounds showed cytotoxic activity and selectivity. The new compound, 3-acridin-9-ylmethyl-5-(5-bromo-1 H-indol-3-ylmethylene)-thiazolidine-2,4-dione (LPSF/AA29 – 7a), proved to be the most promising compound as it presents lower half-maximal inhibitory concentration (IC50) values (ranging from 0.25 to 68.03 µM) depending on cell lineage. In HepG2 cells, the lowest IC50 value was exhibited by 3-acridin-9-ylmethyl-5-(4-piperidin-1-yl-benzylidene)-thiazolidine-2,4-dione (LPSF/AA36 – 7b; 46.95 µM). None of the synthesized compounds showed cytotoxic activity against normal cells (IC50 > 100 µM). The mechanism of death induction and cell cycle effects was also evaluated. Flow cytometric analysis revealed that the compounds LPSF/AA29 – 7a and LPSF/AA36 – 7b significantly increased the percentage of apoptotic cells and induced G2/M arrest in the cell cycle progression. Therefore, these new thiazacridine derivatives constitute promising antitumor agents whose cytotoxicity and selectivity properties indicate they have potential to contribute to or serve as a basis for the development of new cancer drugs in the future.
Collapse
Affiliation(s)
- MBO Chagas
- Laboratory for Immunomodulation and New Therapeutic Approaches, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - NCC Cordeiro
- Laboratory for Planning and Drug Synthesis, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - KMR Marques
- Laboratory for Immunomodulation and New Therapeutic Approaches, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - MG Rocha Pitta
- Laboratory for Planning and Drug Synthesis, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - MJBM Rêgo
- Laboratory for Immunomodulation and New Therapeutic Approaches, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - MCA Lima
- Laboratory for Planning and Drug Synthesis, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - MGR Pitta
- Laboratory for Immunomodulation and New Therapeutic Approaches, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| | - IR Pitta
- Laboratory for Immunomodulation and New Therapeutic Approaches, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
- Laboratory for Planning and Drug Synthesis, Federal University of Pernambuco (UFPE), Recife, PE, Brazil
| |
Collapse
|
49
|
Amsacrine-induced apoptosis of human leukemia U937 cells is mediated by the inhibition of AKT- and ERK-induced stabilization of MCL1. Apoptosis 2016; 22:406-420. [DOI: 10.1007/s10495-016-1307-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
50
|
Graciotti M, Fang Z, Johnsson K, Gönczy P. Chemical Genetic Screen Identifies Natural Products that Modulate Centriole Number. Chembiochem 2016; 17:2063-2074. [DOI: 10.1002/cbic.201600327] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Michele Graciotti
- Institute of Chemical Sciences and Engineering; Swiss Federal Institute of Technology Lausanne (EPFL); 1015 Lausanne Switzerland
- National Centre of Competence in Research (NCCR) in Chemical Biology; 1015 Lausanne Switzerland
| | - Zhou Fang
- Swiss Institute for Experimental Cancer Research (ISREC); Swiss Federal Institute of Technology Lausanne (EPFL); 1015 Lausanne Switzerland
| | - Kai Johnsson
- Institute of Chemical Sciences and Engineering; Swiss Federal Institute of Technology Lausanne (EPFL); 1015 Lausanne Switzerland
- National Centre of Competence in Research (NCCR) in Chemical Biology; 1015 Lausanne Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC); Swiss Federal Institute of Technology Lausanne (EPFL); 1015 Lausanne Switzerland
- National Centre of Competence in Research (NCCR) in Chemical Biology; 1015 Lausanne Switzerland
| |
Collapse
|