1
|
Tang H, Guo X, Yu W, Gao J, Zhu X, Huang Z, Ou W, Zhang H, Chen L, Chen J. Ruthenium(II) complexes as mitochondrial inhibitors of topoisomerase induced A549 cell apoptosis. J Inorg Biochem 2023; 246:112295. [PMID: 37348172 DOI: 10.1016/j.jinorgbio.2023.112295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/27/2023] [Accepted: 06/14/2023] [Indexed: 06/24/2023]
Abstract
Two new ruthenium(II) complexes [Ru(dip)2(PPβC)]PF6 (Ru1, dip = 4,7-diphenyl-1,10-phenanthroline, PPβC = N-(1,10-phenanthrolin-5-yl)-1-phenyl-9H-pyrido[3,4-b]indole-3-carboxamide) and [Ru(phen)2(PPβC)]PF6 (Ru2, phen = 1, 10-phenanthroline) with β-carboline derivative PPβC as the primary ligand, were designed and synthesized. Ru1 and Ru2 displayed higher antiproliferative activity than cisplatin against the test cancer cells, with IC50 values ranging from 0.5 to 3.6 μM. Moreover, Ru1 and Ru2 preferentially accumulated in mitochondria and caused a series of changes in mitochondrial events, including the depolarization of mitochondrial membrane potential, the damage of mitochondrial DNA, the depletion of cellular ATP, and the elevation of intracellular reactive oxygen species levels. Then, it induced caspase-3/7-mediated A549 cell apoptosis. More importantly, both complexes could act as topoisomerase I catalytic inhibitors to inhibit mitochondrial DNA synthesis. Accordingly, the developed Ru(II) complexes hold great potential to be developed as novel therapeutics for cancer treatment.
Collapse
Affiliation(s)
- Hong Tang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, PR China
| | - Xinhua Guo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China
| | - Wenzhu Yu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, PR China
| | - Jie Gao
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, PR China
| | - Xufeng Zhu
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, PR China
| | - Zunnan Huang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; Key Laboratory of Computer-Aided Drug Design of Dongguan City, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China
| | - Wenhui Ou
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China
| | - Hanfu Zhang
- School of Molecular Science, The University of Western Australia, Perth 6009, WA, Australia
| | - Lanmei Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; Key Laboratory of Computer-Aided Drug Design of Dongguan City, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China.
| | - Jincan Chen
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, PR China; Key Laboratory of Computer-Aided Drug Design of Dongguan City, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, PR China.
| |
Collapse
|
2
|
Jian JY, McCarty KD, Byl J, Guengerich FP, Neuman K, Osheroff N. Basis for the discrimination of supercoil handedness during DNA cleavage by human and bacterial type II topoisomerases. Nucleic Acids Res 2023; 51:3888-3902. [PMID: 36999602 PMCID: PMC10164583 DOI: 10.1093/nar/gkad190] [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: 12/05/2022] [Revised: 02/24/2023] [Accepted: 03/23/2023] [Indexed: 04/01/2023] Open
Abstract
To perform double-stranded DNA passage, type II topoisomerases generate a covalent enzyme-cleaved DNA complex (i.e. cleavage complex). Although this complex is a requisite enzyme intermediate, it is also intrinsically dangerous to genomic stability. Consequently, cleavage complexes are the targets for several clinically relevant anticancer and antibacterial drugs. Human topoisomerase IIα and IIβ and bacterial gyrase maintain higher levels of cleavage complexes with negatively supercoiled over positively supercoiled DNA substrates. Conversely, bacterial topoisomerase IV is less able to distinguish DNA supercoil handedness. Despite the importance of supercoil geometry to the activities of type II topoisomerases, the basis for supercoil handedness recognition during DNA cleavage has not been characterized. Based on the results of benchtop and rapid-quench flow kinetics experiments, the forward rate of cleavage is the determining factor of how topoisomerase IIα/IIβ, gyrase and topoisomerase IV distinguish supercoil handedness in the absence or presence of anticancer/antibacterial drugs. In the presence of drugs, this ability can be enhanced by the formation of more stable cleavage complexes with negatively supercoiled DNA. Finally, rates of enzyme-mediated DNA ligation do not contribute to the recognition of DNA supercoil geometry during cleavage. Our results provide greater insight into how type II topoisomerases recognize their DNA substrates.
Collapse
Affiliation(s)
- Jeffrey Y Jian
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kevin D McCarty
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jo Ann W Byl
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Keir C Neuman
- Laboratory of Single Molecule Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20982, 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
|
3
|
Olatunde OZ, Yong J, Lu C, Ming Y. A Review on Shikonin and Its Derivatives as Potent Anticancer Agents Targeted against Topoisomerases. Curr Med Chem 2023; 31:CMC-EPUB-129356. [PMID: 36752292 DOI: 10.2174/0929867330666230208094828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/12/2022] [Accepted: 11/30/2022] [Indexed: 02/09/2023]
Abstract
The topoisomerases (TOPO) play indispensable roles in DNA metabolism, by regulating the topological state of DNA. Topoisomerase I and II are the well-established drug-targets for the development of anticancer agents and antibiotics. These drugs-targeting enzymes have been used to establish the relationship between drug-stimulated DNA cleavable complex formation and cytotoxicity. Some anticancer drugs (such as camptothecin, anthracyclines, mitoxantrone) are also widely used as Topo I and Topo II inhibitors, but the poor water solubility, myeloma suppression, dose-dependent cardiotoxicity, and multidrug resistance (MDR) limited their prolong use as therapeutics. Also, most of these agents displayed selective inhibition only against Topo I or II. In recent years, researchers focus on the design and synthesis of the dual Topo I and II inhibitors, or the discovery of the dual Topo I and II inhibitors from natural products. Shikonin (a natural compound with anthraquinone skeleton, isolated from the roots of Lithospermum erythrorhizon) has drawn much attention due to its wide spectrum of anticancer activities, especially due to its dual Topo inhibitive performance, and without the adverse side effects, and different kinds of shikonin derivatives have been synthesized as TOPO inhibitors for the development of anticancer agents. In this review, the progress of the shikonin and its derivatives together with their anticancer activities, anticancer mechanism, and their structure-activity relationship (SAR) was comprehensively summarized by searching the CNKI, PubMed, Web of Science, Scopus, and Google Scholar databases.
Collapse
Affiliation(s)
- Olagoke Zacchaeus Olatunde
- Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian,350002, China
| | - Jianping Yong
- Xiamen Institute of Rare-earth Materials, Chinese Academy of Sciences, Xiamen, Fujian, 361021, China
| | - Canzhong Lu
- Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian,350002, China
- Xiamen Institute of Rare-earth Materials, Chinese Academy of Sciences, Xiamen, Fujian, 361021, China
| | - Yanlin Ming
- Fujian Institute of Subtropical Botany, Xiamen, Fujian, 361006, China
| |
Collapse
|
4
|
Hwang SY, Shrestha A, Park S, Bist G, Kunwar S, Kadayat TM, Jang H, Seo M, Sheen N, Kim S, Jeon KH, Lee ES, Kwon Y. Identification of new halogen-containing 2,4-diphenyl indenopyridin-5-one derivative as a boosting agent for the anticancer responses of clinically available topoisomerase inhibitors. Eur J Med Chem 2022; 227:113916. [PMID: 34678573 DOI: 10.1016/j.ejmech.2021.113916] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022]
Abstract
Based on previous reports on the significance of halogen moieties and the indenopyridin-5-one skeleton, we designed and synthesized a novel series of halogen (F-, Cl-, Br-, CF3- and OCF3-)-containing 2,4-diphenyl indenopyridin-5-ones and their corresponding -5-ols. Unlike indenopyridin-5-ols, most of the prepared indenopyridin-5-ones with Cl-, Br-, and CF3- groups at the 2-phenyl ring conferred a strong dual topoisomerase I/IIα inhibitory effect. Among the series, para-bromophenyl substituted compound 9 exhibited the most potent topoisomerase inhibition and antiproliferative effects, which showed dependency upon the topoisomerase gene expression level of diverse cancer cells. In particular, as a DNA minor groove-binding non-intercalative topoisomerase I/IIα catalytic inhibitor, compound 9 synergistically promoted the anticancer efficacy of clinically applied topoisomerase I/IIα poisons both in vitro and in vivo, having the great advantage of alleviating poison-related toxicities.
Collapse
Affiliation(s)
- Soo-Yeon Hwang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Aarajana Shrestha
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Seojeong Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Ganesh Bist
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Surendra Kunwar
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Tara Man Kadayat
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Haejin Jang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Minjung Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Naeun Sheen
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seojeong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Kyung-Hwa Jeon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Eung-Seok Lee
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| | - Youngjoo Kwon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
5
|
9-Bromo-2,3-diethylbenzo[de]chromene-7,8-dione (MSN54): A novel non-intercalative topoisomerase II catalytic inhibitor. Bioorg Chem 2021; 114:105097. [PMID: 34171594 DOI: 10.1016/j.bioorg.2021.105097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 05/30/2021] [Accepted: 06/10/2021] [Indexed: 11/22/2022]
Abstract
Novel mansonone F derivative MSN54 (9-bromo-2,3-diethylbenzo[de]chromene-7,8-dione) exhibited significant cytotoxicity against twelve human tumor cell lines in vitro, with particularly strong potency against HL-60/MX2 cell line resistant to Topo II poisons. MSN54 was found to have IC50 of 0.69 and 1.43 µM against HL-60 and HL-60/MX2 cells, respectively. The resistance index is 10 times lower than that of the positive control VP-16 (etoposide). Various biological assays confirmed that MSN54 acted as a Topo IIα specific non-intercalative catalytic inhibitor. Furthermore, MSN54 exhibited good antitumor efficacy and low toxicity at a dose of 5 mg/kg in A549 tumor xenograft models. Thus, compound MSN54 is a promising candidate for the development of novel antitumor agents.
Collapse
|
6
|
Kang M, Fu R, Zhang P, Lou S, Yang X, Chen Y, Ma T, Zhang Y, Xi Z, Liu J. A chromosome-level Camptotheca acuminata genome assembly provides insights into the evolutionary origin of camptothecin biosynthesis. Nat Commun 2021; 12:3531. [PMID: 34112794 PMCID: PMC8192753 DOI: 10.1038/s41467-021-23872-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 05/20/2021] [Indexed: 02/05/2023] Open
Abstract
Camptothecin and its derivatives are widely used for treating malignant tumors. Previous studies revealed only a limited number of candidate genes for camptothecin biosynthesis in Camptotheca acuminata, and it is still poorly understood how its biosynthesis of camptothecin has evolved. Here, we report a high-quality, chromosome-level C. acuminata genome assembly. We find that C. acuminata experiences an independent whole-genome duplication and numerous genes derive from it are related to camptothecin biosynthesis. Comparing with Catharanthus roseus, the loganic acid O-methyltransferase (LAMT) in C. acuminata fails to convert loganic acid into loganin. Instead, two secologanic acid synthases (SLASs) convert loganic acid to secologanic acid. The functional divergence of the LAMT gene and positive evolution of two SLAS genes, therefore, both contribute greatly to the camptothecin biosynthesis in C. acuminata. Our results emphasize the importance of high-quality genome assembly in identifying genetic changes in the evolutionary origin of a secondary metabolite.
Collapse
Affiliation(s)
- Minghui Kang
- Key Laboratory of Bio-resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Rao Fu
- Key Laboratory of Bio-resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Pingyu Zhang
- Key Laboratory of Bio-resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Shangling Lou
- Key Laboratory of Bio-resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Xuchen Yang
- Key Laboratory of Bio-resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yang Chen
- Key Laboratory of Bio-resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Tao Ma
- Key Laboratory of Bio-resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yang Zhang
- Key Laboratory of Bio-resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Zhenxiang Xi
- Key Laboratory of Bio-resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jianquan Liu
- Key Laboratory of Bio-resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
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: 4.3] [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
|
8
|
Ortega JA, Arencibia JM, Minniti E, Byl JAW, Franco-Ulloa S, Borgogno M, Genna V, Summa M, Bertozzi SM, Bertorelli R, Armirotti A, Minarini A, Sissi C, Osheroff N, De Vivo M. Novel, Potent, and Druglike Tetrahydroquinazoline Inhibitor That Is Highly Selective for Human Topoisomerase II α over β. J Med Chem 2020; 63:12873-12886. [PMID: 33079544 PMCID: PMC7668297 DOI: 10.1021/acs.jmedchem.0c00774] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
We disclose a novel
class of 6-amino-tetrahydroquinazoline derivatives
that inhibit human topoisomerase II (topoII), a validated target of
anticancer drugs. In contrast to topoII-targeted drugs currently in
clinical use, these compounds do not act as topoII poisons that enhance
enzyme-mediated DNA cleavage, a mechanism that is linked to the development
of secondary leukemias. Instead, these tetrahydroquinazolines block
the topoII function with no evidence of DNA intercalation. We identified
a potent lead compound [compound 14 (ARN-21934) IC50 = 2 μM for inhibition of DNA relaxation, as compared
to an IC50 = 120 μM for the anticancer drug etoposide]
with excellent metabolic stability and solubility. This new compound
also shows ~100-fold selectivity for topoIIα over topoβ,
a broad antiproliferative activity toward cultured human cancer cells,
a favorable in vivo pharmacokinetic profile, and the ability to penetrate
the blood–brain barrier. Thus, ARN-21934 is a highly promising
lead for the development of novel and potentially safer topoII-targeted
anticancer drugs.
Collapse
Affiliation(s)
- Jose Antonio Ortega
- Laboratory of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Jose M Arencibia
- Laboratory of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Elirosa Minniti
- Laboratory of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy.,Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Jo Ann W Byl
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | - Sebastian Franco-Ulloa
- Laboratory of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Marco Borgogno
- Laboratory of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Vito Genna
- Laboratory of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Maria Summa
- Analytical Chemistry & Translational Pharmacology, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Sine Mandrup Bertozzi
- Analytical Chemistry & Translational Pharmacology, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Rosalia Bertorelli
- Analytical Chemistry & Translational Pharmacology, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Andrea Armirotti
- Analytical Chemistry & Translational Pharmacology, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Anna Minarini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Claudia Sissi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Neil Osheroff
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States.,Department of Medicine (Hematology/Oncology), Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6307, United States.,VA Tennessee Valley Healthcare System, Nashville, Tennessee 37212, United States
| | - Marco De Vivo
- Laboratory of Molecular Modeling and Drug Discovery, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| |
Collapse
|
9
|
Gibson EG, Oviatt AA, Osheroff N. Two-Dimensional Gel Electrophoresis to Resolve DNA Topoisomers. Methods Mol Biol 2020; 2119:15-24. [PMID: 31989511 PMCID: PMC7012906 DOI: 10.1007/978-1-0716-0323-9_2] [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] [Indexed: 01/19/2023]
Abstract
Agarose gel electrophoresis is one of the most straightforward techniques that can be used to differentiate between topoisomers of closed circular DNA molecules. Generally, the products of reactions that monitor the interconversion of DNA between negatively supercoiled and relaxed DNA or positively supercoiled and relaxed DNA can be resolved by one-dimensional gel electrophoresis. However, in more complex reactions that contain both positively and negatively supercoiled DNA, one-dimensional resolution is insufficient. In these cases, a second dimension of gel electrophoresis is necessary. This chapter describes the technique of two-dimensional agarose gel electrophoresis and how it can be used to resolve a spectrum of DNA topoisomers.
Collapse
Affiliation(s)
- Elizabeth G. Gibson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Alexandria A. Oviatt
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Neil Osheroff
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States,,Department of Medicine (Hematology/Oncology), Vanderbilt University School of Medicine, Nashville, TN 37232, United States,,VA Tennessee Valley Healthcare System, Nashville, TN 37212, United States
| |
Collapse
|
10
|
Yousuf I, Arjmand F, Tabassum S, Ahmad M. Design and synthesis of a DNA intercalative half-sandwich organoruthenium( ii)–chromone complex: cytotoxicity evaluation and topoisomerase Iα inhibition assay. NEW J CHEM 2019; 43:5475-5487. [DOI: 10.1039/c9nj00042a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
A half-sandwich organoruthenium(ii)–chromone complex acts as a potential topoisomerase I inhibitor.
Collapse
Affiliation(s)
- Imtiyaz Yousuf
- Department of Chemistry, Aligarh Muslim University
- Aligarh 202002
- India
| | - Farukh Arjmand
- Department of Chemistry, Aligarh Muslim University
- Aligarh 202002
- India
| | - Sartaj Tabassum
- Department of Chemistry, Aligarh Muslim University
- Aligarh 202002
- India
| | - Musheer Ahmad
- Department of Applied Chemistry, Aligarh Muslim University
- Aligarh 202002
- India
| |
Collapse
|
11
|
Arepalli SK, Lee C, Sim S, Lee K, Jo H, Jun KY, Kwon Y, Kang JS, Jung JK, Lee H. Development of 13H-benzo[f]chromeno[4,3-b][1,7]naphthyridines and their salts as potent cytotoxic agents and topoisomerase I/IIα inhibitors. Bioorg Med Chem 2018; 26:5181-5193. [PMID: 30253887 DOI: 10.1016/j.bmc.2018.09.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/27/2018] [Accepted: 09/17/2018] [Indexed: 10/28/2022]
Abstract
A novel series of 35 angularly fused pentacyclic 13H-benzo[f]chromeno[4,3-b][1,7]naphthyridines and 13H-benzo[f]chromeno[4,3-b][1,7]naphthyridin-5-ium chlorides were designed and synthesized. Their cytotoxic activities were investigated against six human cancer cell lines (NCIH23, HCT15, NUGC-3, ACHN, PC-3, and MDA-MB-231). Among all screened compounds; 28, 30, 34, 35, 46, 48, 52, and 53 compounds exhibited potent cytotoxic activities against all tested human cancer cell lines. Further, these potent lead cytotoxic agents were evaluated against human Topoisomerase I and IIα inhibition. Among them, the compound 48 exhibited dual Topoisomerase I and IIα inhibition especially at 20 μM concentrations the compound 48 exhibited 1.25 times more potent Topoisomerase IIα inhibitory activity (38.3%) than the reference drug etoposide (30.6%). The compound 52 also exhibited excellent (88.4%) topoisomerase I inhibition than the reference drug camptothecin (66.7%) at 100 μM concentrations. Molecular docking studies of the compounds 48 and 52 with topo I discovered that they both intercalated into the DNA single-strand cleavage site where the compound 48 have van der Waals interactions with residues Arg364, Pro431, and Asn722 whilst the compound 52 have with Arg364, Thr718, and Asn722 residues. Both the compounds 48 and 52 have π-π stacking interactions with the stacked DNA bases. The docking studies of the compound 48 with topo IIα explored that it was bound to the topo IIα DNA cleavage site where etoposide was situated. The benzo[f]chromeno[4,3-b][1,7]naphthyridine ring of the compound 48 was stacked between the DNA bases of the cleavage site with π-π stacking interactions and there were no hydrogen bond interactions with topo IIα.
Collapse
Affiliation(s)
| | - Chaerim Lee
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Seongrak Sim
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Kiho Lee
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Hyunji Jo
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Kyu-Yeon Jun
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Youngjoo Kwon
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jong-Soon Kang
- Korea Research Institute of Bioscience and Biotechnology, Chungbuk 28116, Republic of Korea
| | - Jae-Kyung Jung
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Heesoon Lee
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea.
| |
Collapse
|
12
|
Afzal M, Al-Lohedan HA, Usman M, Tabassum S. Carbohydrate-based heteronuclear complexes as topoisomerase Iα inhibitor: approach toward anticancer chemotherapeutics. J Biomol Struct Dyn 2018; 37:1494-1510. [PMID: 29606083 DOI: 10.1080/07391102.2018.1459321] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Due to the critical role of cellular enzymes necessary for cell proliferation by deciphering topological hurdles in the process of DNA replication, topoisomerases have been one of the major targets in the anticancer drug development area. A need, therefore, arises for new metallodrugs that specifically recognizes DNA and inhibits the activity of topoisomerase enzymes, herein, we report the synthesis and characterization of new metal-based glycoconjugate entities containing heterobimetallic core CuII-SnIV (1) and NiII-SnIV (2) derived from N-glycoside ligand (L). The optimized structure of complex 1 and other significant vibrational modes have been explained using dispersion corrected B3LYP/DFT calculations. In vitro DNA binding profile of the L and both the complexes 1 and 2 were done by various biophysical studies. Complex 1 breaks pBR322 DNA via a hydrolytic means which was validated by T4 DNA enzymatic assay. To get a mechanistic insight of mode of action topoisomerase I (Topo I) inhibition assay was carried out. Also, we have taken the help of molecular modeling studies in accordance with experimental findings. In vitro cytotoxicity of the complex 1 was evaluated against a panel of cancer cells which exhibited remarkably good anticancer activity (GI50 values <10 μg/ml). Moreover, intracellular localization of the complex 1 was visualized by confocal microscopy against HeLa cells.
Collapse
Affiliation(s)
- Mohd Afzal
- a Department of Chemistry , Aligarh Muslim University , Aligarh 202002 , India
| | - Hamad A Al-Lohedan
- b Surfactant Research Chair, Department of Chemistry, College of Science , King Saud University , P.O. Box 2455, Riyadh 11451 , Saudi Arabia
| | - Mohammad Usman
- a Department of Chemistry , Aligarh Muslim University , Aligarh 202002 , India
| | - Sartaj Tabassum
- b Surfactant Research Chair, Department of Chemistry, College of Science , King Saud University , P.O. Box 2455, Riyadh 11451 , Saudi Arabia
| |
Collapse
|
13
|
Yousuf I, Usman M, Ahmad M, Tabassum S, Arjmand F. Single X-ray crystal structure, DFT studies and topoisomerase I inhibition activity of a tailored ionic Ag( i) nalidixic acid–piperazinium drug entity specific for pancreatic cancer cells. NEW J CHEM 2018; 42:506-519. [DOI: 10.1039/c7nj03602g] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
DFT studies, Topo I inhibition assay and cytotoxic activity of novel ionic Ag(i) nalidixic acid–piperazinium molecular entity.
Collapse
Affiliation(s)
- Imtiyaz Yousuf
- Department of Chemistry
- Aligarh Muslim University
- Aligarh 202002
- India
| | - Mohammad Usman
- Department of Chemistry
- Aligarh Muslim University
- Aligarh 202002
- India
| | - Musheer Ahmad
- Department of Applied Chemistry
- Aligarh Muslim University
- Aligarh 202002
- India
| | - Sartaj Tabassum
- Department of Chemistry
- Aligarh Muslim University
- Aligarh 202002
- India
| | - Farukh Arjmand
- Department of Chemistry
- Aligarh Muslim University
- Aligarh 202002
- India
| |
Collapse
|
14
|
Bist G, Park S, Song C, Thapa Magar TB, Shrestha A, Kwon Y, Lee ES. Dihydroxylated 2,6-diphenyl-4-chlorophenylpyridines: Topoisomerase I and IIα dual inhibitors with DNA non-intercalative catalytic activity. Eur J Med Chem 2017; 133:69-84. [DOI: 10.1016/j.ejmech.2017.03.048] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 01/10/2023]
|
15
|
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: 2.9] [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
|
16
|
van Gijn R, Lendfers RRH, Schellens JHM, Bult A, Beijnen JH. Dual topoisomerase I/II inhibitors. J Oncol Pharm Pract 2016. [DOI: 10.1177/107815520000600303] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Topoisomerase (topo) I and II are nuclear enzymes, which play a major role in the topological rearrangement of DNA during replication and transcription processes. In the course of years, many different agents have been found which can inhibit the topos and thereby exploit cytotoxicity, also against tumour cells. Selective inhibition of the topo I enzyme can, however, induce a reactive increase in topo II levels, and vice versa. This mechanism is associated with the development of drug resistance. Dual inhibition of both topo I and II may, theoretically, overcome this resistance problem. In this review, the most important and promising dual topo I/II inhibitors designed as anticancer agents will be discussed. Thus far, only the indolyl quinoline derivative TAS-103, the 7 H-benzo [ e] pyrido [4,3- b] indole derivative intoplicine, and the acridine derivative PZA have been shown to be dual topo inhibitors with high cytotoxicity.
Collapse
Affiliation(s)
- R van Gijn
- Department of Pharmacy and Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - RRH Lendfers
- Department of Biomedical Analysis, Faculty of Pharmacy, Utrecht University, Utrecht, The Netherlands
| | - JHM Schellens
- Department of Biomedical Analysis, Faculty of Pharmacy, Utrecht University, Utrecht, The Netherlands, Division of Medical Oncology, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - A Bult
- Department of Biomedical Analysis, Faculty of Pharmacy, Utrecht University, Utrecht, The Netherlands
| | - JH Beijnen
- Department of Pharmacy and Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute, Amsterdam, The Netherlands, Department of Biomedical Analysis, Faculty of Pharmacy, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
17
|
Fluoroquinolone interactions with Mycobacterium tuberculosis gyrase: Enhancing drug activity against wild-type and resistant gyrase. Proc Natl Acad Sci U S A 2016; 113:E839-46. [PMID: 26792518 DOI: 10.1073/pnas.1525055113] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mycobacterium tuberculosis is a significant source of global morbidity and mortality. Moxifloxacin and other fluoroquinolones are important therapeutic agents for the treatment of tuberculosis, particularly multidrug-resistant infections. To guide the development of new quinolone-based agents, it is critical to understand the basis of drug action against M. tuberculosis gyrase and how mutations in the enzyme cause resistance. Therefore, we characterized interactions of fluoroquinolones and related drugs with WT gyrase and enzymes carrying mutations at GyrA(A90) and GyrA(D94). M. tuberculosis gyrase lacks a conserved serine that anchors a water-metal ion bridge that is critical for quinolone interactions with other bacterial type II topoisomerases. Despite the fact that the serine is replaced by an alanine (i.e., GyrA(A90)) in M. tuberculosis gyrase, the bridge still forms and plays a functional role in mediating quinolone-gyrase interactions. Clinically relevant mutations at GyrA(A90) and GyrA(D94) cause quinolone resistance by disrupting the bridge-enzyme interaction, thereby decreasing drug affinity. Fluoroquinolone activity against WT and resistant enzymes is enhanced by the introduction of specific groups at the C7 and C8 positions. By dissecting fluoroquinolone-enzyme interactions, we determined that an 8-methyl-moxifloxacin derivative induces high levels of stable cleavage complexes with WT gyrase and two common resistant enzymes, GyrA(A90V) and GyrA(D94G). 8-Methyl-moxifloxacin was more potent than moxifloxacin against WT M. tuberculosis gyrase and displayed higher activity against the mutant enzymes than moxifloxacin did against WT gyrase. This chemical biology approach to defining drug-enzyme interactions has the potential to identify novel drugs with improved activity against tuberculosis.
Collapse
|
18
|
Du L, Li MD, Zhang Y, Xue J, Zhang X, Zhu R, Cheng SC, Li X, Phillips DL. Photoconversion of β-Lapachone to α-Lapachone via a Protonation-Assisted Singlet Excited State Pathway in Aqueous Solution: A Time-Resolved Spectroscopic Study. J Org Chem 2015; 80:7340-50. [DOI: 10.1021/acs.joc.5b00086] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Lili Du
- Department of Chemistry, The University of Hong Kong, Hong Kong S.A.R., China
| | - Ming-De Li
- Department of Chemistry, The University of Hong Kong, Hong Kong S.A.R., China
| | - Yanfeng Zhang
- Department of Chemistry, The University of Hong Kong, Hong Kong S.A.R., China
| | - Jiadan Xue
- Department
of Chemistry, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xiting Zhang
- Department of Chemistry, The University of Hong Kong, Hong Kong S.A.R., China
| | - Ruixue Zhu
- Department of Chemistry, The University of Hong Kong, Hong Kong S.A.R., China
| | - Shun Cheung Cheng
- Department of Chemistry, The University of Hong Kong, Hong Kong S.A.R., China
| | - Xuechen Li
- Department of Chemistry, The University of Hong Kong, Hong Kong S.A.R., China
| | - David Lee Phillips
- Department of Chemistry, The University of Hong Kong, Hong Kong S.A.R., China
| |
Collapse
|
19
|
Du K, Liang J, Wang Y, Kou J, Qian C, Ji L, Chao H. Dual inhibition of topoisomerases I and IIα by ruthenium(II) complexes containing asymmetric tridentate ligands. Dalton Trans 2015; 43:17303-16. [PMID: 25315107 DOI: 10.1039/c4dt02142h] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Five novel ruthenium(II) complexes, [Ru(dtzp)(dppt)](2+) (1), [Ru(dtzp)(pti)](2+) (2), [Ru(dtzp)(ptn)](2+) (3), [Ru(dtzp)(pta)](2+) (4) and [Ru(dtzp)(ptp)](2+) (5) (where dtzp = 2,6-di(thiazol-2-yl)pyridine, dppt = 3-(1,10-phenanthroline-2-yl)-5,6-diphenyl-as-triazine), pti = 3-(1,10-phenanthroline-2-yl)-as-triazino-[5,6-f]isatin, ptn = 3-(1,10-phenanthroline-2-yl)-as-triazino[5,6-f]naphthalene, pta = 3-(1,10-phenanthroline-2-yl)-as-triazino[5,6-f]acenaphthylene, and ptp = 3-(1,10-phenanthroline-2-yl)-as-triazino[5,6-f]-phenanthrene), were synthesised and characterised. The structures of complexes 3-5 were determined by X-ray diffraction. The DNA binding behaviours of the complexes were studied by spectroscopic and viscosity measurements. The results suggested that the Ru(II) complexes, except for complex 1, bind to DNA in an intercalative mode. Topoisomerase inhibition and DNA strand passage assay confirmed that Ru(II) complexes 3, 4, and 5 acted as efficient dual inhibitors of topoisomerases I and IIα. In vitro cytotoxicity assays indicated that these complexes exhibited anticancer activity against various cancer cell lines. Ruthenium(ii) complexes were confirmed to preferentially accumulate in the nucleus of cancer cells and induced DNA damage. Flow cytometric analysis and AO/EB staining assays indicated that these complexes induced cell apoptosis. With the loss of the mitochondrial membrane potential, the Ru(ii) complexes induce apoptosis via the mitochondrial pathway.
Collapse
Affiliation(s)
- Kejie Du
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, State Key Laboratory of Optoelectronic Materials and Technologies, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
20
|
Design, synthesis and biological evaluation of novel 7-alkylamino substituted benzo[a]phenazin derivatives as dual topoisomerase I/II inhibitors. Eur J Med Chem 2015; 92:540-53. [DOI: 10.1016/j.ejmech.2015.01.024] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 01/10/2015] [Accepted: 01/11/2015] [Indexed: 11/22/2022]
|
21
|
Mokesch S, Novak MS, Roller A, Jakupec MA, Kandioller W, Keppler BK. 1,3-Dioxoindan-2-carboxamides as Bioactive Ligand Scaffolds for the Development of Novel Organometallic Anticancer Drugs. Organometallics 2015. [DOI: 10.1021/om501032s] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Stephan Mokesch
- Institute of Inorganic Chemistry, Faculty of Chemistry, and ‡Research
Platform “Translational Cancer Therapy Research”, University of Vienna, Waehringer Straße 42, A-1090 Vienna, Austria
| | - Maria S. Novak
- Institute of Inorganic Chemistry, Faculty of Chemistry, and ‡Research
Platform “Translational Cancer Therapy Research”, University of Vienna, Waehringer Straße 42, A-1090 Vienna, Austria
| | - Alexander Roller
- Institute of Inorganic Chemistry, Faculty of Chemistry, and ‡Research
Platform “Translational Cancer Therapy Research”, University of Vienna, Waehringer Straße 42, A-1090 Vienna, Austria
| | - Michael A. Jakupec
- Institute of Inorganic Chemistry, Faculty of Chemistry, and ‡Research
Platform “Translational Cancer Therapy Research”, University of Vienna, Waehringer Straße 42, A-1090 Vienna, Austria
| | - Wolfgang Kandioller
- Institute of Inorganic Chemistry, Faculty of Chemistry, and ‡Research
Platform “Translational Cancer Therapy Research”, University of Vienna, Waehringer Straße 42, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- Institute of Inorganic Chemistry, Faculty of Chemistry, and ‡Research
Platform “Translational Cancer Therapy Research”, University of Vienna, Waehringer Straße 42, A-1090 Vienna, Austria
| |
Collapse
|
22
|
Aldred KJ, Breland EJ, Vlčková V, Strub MP, Neuman KC, Kerns RJ, Osheroff N. Role of the water-metal ion bridge in mediating interactions between quinolones and Escherichia coli topoisomerase IV. Biochemistry 2014; 53:5558-67. [PMID: 25115926 PMCID: PMC4151693 DOI: 10.1021/bi500682e] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
![]()
Although
quinolones have been in clinical use for decades, the
mechanism underlying drug activity and resistance has remained elusive.
However, recent studies indicate that clinically relevant quinolones
interact with Bacillus anthracis (Gram-positive)
topoisomerase IV through a critical water–metal ion bridge
and that the most common quinolone resistance mutations decrease drug
activity by disrupting this bridge. As a first step toward determining
whether the water–metal ion bridge is a general mechanism of
quinolone–topoisomerase interaction, we characterized drug
interactions with wild-type Escherichia coli (Gram-negative)
topoisomerase IV and a series of ParC enzymes with mutations (S80L,
S80I, S80F, and E84K) in the predicted bridge-anchoring residues.
Results strongly suggest that the water–metal ion bridge is
essential for quinolone activity against E. coli topoisomerase
IV. Although the bridge represents a common and critical mechanism
that underlies broad-spectrum quinolone function, it appears to play
different roles in B. anthracis and E. coli topoisomerase IV. The water–metal ion bridge is the most
important binding contact of clinically relevant quinolones with the
Gram-positive enzyme. However, it primarily acts to properly align
clinically relevant quinolones with E. coli topoisomerase
IV. Finally, even though ciprofloxacin is unable to increase levels
of DNA cleavage mediated by several of the Ser80 and Glu84 mutant E. coli enzymes, the drug still retains the ability to inhibit
the overall catalytic activity of these topoisomerase IV proteins.
Inhibition parallels drug binding, suggesting that the presence of
the drug in the active site is sufficient to diminish DNA relaxation
rates.
Collapse
Affiliation(s)
- Katie J Aldred
- Department of Biochemistry and ‡Department of Medicine (Hematology/Oncology), Vanderbilt University School of Medicine , Nashville, Tennessee 37232-0146, United States
| | | | | | | | | | | | | |
Collapse
|
23
|
Synthesis and characterization of Cu(II)-based anticancer chemotherapeutic agent targeting topoisomerase Iα: In vitro DNA binding, pBR322 cleavage, molecular docking studies and cytotoxicity against human cancer cell lines. Eur J Med Chem 2014; 74:509-23. [PMID: 24508781 DOI: 10.1016/j.ejmech.2013.12.046] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/23/2013] [Accepted: 12/26/2013] [Indexed: 02/07/2023]
|
24
|
Zhuo ST, Li CY, Hu MH, Chen SB, Yao PF, Huang SL, Ou TM, Tan JH, An LK, Li D, Gu LQ, Huang ZS. Synthesis and biological evaluation of benzo[a]phenazine derivatives as a dual inhibitor of topoisomerase I and II. Org Biomol Chem 2014; 11:3989-4005. [PMID: 23657605 DOI: 10.1039/c3ob40325d] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Topoisomerases (Topo I and Topo II) are very important players in DNA replication, repair, and transcription, and are a promising class of antitumor target. In present study, a series of benzo[a]phenazine derivatives with alkylamino side chains at C-5 were designed, synthesized, and their biological activities were evaluated. Most of derivatives showed good antiproliferative activity with a range of IC50 values of 1-10 μM on the four cancer cell lines HeLa, A549, MCF-7, and HL-60. Topoisomerase-mediated DNA relaxation assay results showed that derivatives could effectively inhibit the activity of both Topo I and Topo II, and the structure-activity relationship studies indicated the importance of introducing an alkylamino side chain. Further mechanism studies revealed that the compounds could stabilize the Topo I-DNA cleavage complexes and inhibit the ATPase activity of hTopo II, indicating that they are a rare class of dual topoisomerase inhibitors by acting as Topo I poisons and Topo II catalytic inhibitors. Moreover, flow cytometric analysis and caspase-3/7 activation assay showed that this class of compounds could induce apoptosis of HL-60 cells.
Collapse
Affiliation(s)
- Shi-Tian Zhuo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
He X, Zeng L, Yang G, Xie L, Sun X, Tan L. DNA binding, photocleavage and topoisomerase inhibitory activity of polypyridyl ruthenium(II) complexes containing the same ancillary ligand and different main ligands. Inorganica Chim Acta 2013. [DOI: 10.1016/j.ica.2013.08.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
26
|
Huang ZH, Zhuo ST, Li CY, Xie HT, Li D, Tan JH, Ou TM, Huang ZS, Gu LQ, Huang SL. Design, synthesis and biological evaluation of novel mansonone E derivatives prepared via CuAAC click chemistry as topoisomerase II inhibitors. Eur J Med Chem 2013; 68:58-71. [DOI: 10.1016/j.ejmech.2013.07.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 07/18/2013] [Accepted: 07/24/2013] [Indexed: 11/26/2022]
|
27
|
Ruthenium (II) complexes containing a new asymmetric ligand: DNA interaction, photocleavage and topoisomerase I inhibition. J Organomet Chem 2013. [DOI: 10.1016/j.jorganchem.2013.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
28
|
Aldred KJ, McPherson SA, Turnbough CL, Kerns RJ, Osheroff N. Topoisomerase IV-quinolone interactions are mediated through a water-metal ion bridge: mechanistic basis of quinolone resistance. Nucleic Acids Res 2013; 41:4628-39. [PMID: 23460203 PMCID: PMC3632122 DOI: 10.1093/nar/gkt124] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although quinolones are the most commonly prescribed antibacterials, their use is threatened by an increasing prevalence of resistance. The most common causes of quinolone resistance are mutations of a specific serine or acidic residue in the A subunit of gyrase or topoisomerase IV. These amino acids are proposed to serve as a critical enzyme-quinolone interaction site by anchoring a water-metal ion bridge that coordinates drug binding. To probe the role of the proposed water-metal ion bridge, we characterized wild-type, GrlAE85K, GrlAS81F/E85K, GrlAE85A, GrlAS81F/E85A and GrlAS81FBacillus anthracis topoisomerase IV, their sensitivity to quinolones and related drugs and their use of metal ions. Mutations increased the Mg2+ concentration required to produce maximal quinolone-induced DNA cleavage and restricted the divalent metal ions that could support quinolone activity. Individual mutation of Ser81 or Glu85 partially disrupted bridge function, whereas simultaneous mutation of both residues abrogated protein–quinolone interactions. Results provide functional evidence for the existence of the water-metal ion bridge, confirm that the serine and glutamic acid residues anchor the bridge, demonstrate that the bridge is the primary conduit for interactions between clinically relevant quinolones and topoisomerase IV and provide a likely mechanism for the most common causes of quinolone resistance.
Collapse
Affiliation(s)
- Katie J Aldred
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146, USA
| | | | | | | | | |
Collapse
|
29
|
He X, Yang G, Sun X, Xie L, Tan L. Synthesis and Characterisation of RuII Polypyridyl Complexes: DNA-Binding, Photocleavage, and Topoisomerase I and II Inhibitory Activity. Aust J Chem 2013. [DOI: 10.1071/ch13329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Two mixed-ligand ruthenium(ii) complexes [Ru(phen)2(cptcp)]2+ (Ru1; phen = 1,10-phenanthroline, cptcp = 2-(4-carbazol-9-yl-phenyl)-1H-1,3,7,8-tetraaza-cyclopenta-[l]-phenanthrene) and [Ru(phen)2(btcpc)]2+ (Ru2; btcpc = 9-butyl-6-(1H-1,3,7,8-tetraaza-cyclo-cyclopenta-[l]-phenanthren-2-yl)-9H-carbazole-3-carbaldehyde) have been synthesised and characterised. The DNA-binding behaviours of the two complexes have been investigated by using spectroscopic and viscosity measurements. Results suggest that the two complexes bind to DNA by intercalation. The photocleavage of plasmid pBR322 DNA indicates that Ru1 exhibits more effective DNA cleavage activity in comparison to that exhibited by Ru2 under the same conditions, and different cleavage mechanisms are determined. Topoisomerase inhibition and DNA strand passage assay confirm that Ru1 may act as an efficient dual inhibitor of topoisomerases I and II, whereas Ru2 may only act as a single inhibitor of topoisomerases II.
Collapse
|
30
|
Cinelli MA, Reddy PN, Lv PC, Liang JH, Chen L, Agama K, Pommier Y, van Breemen RB, Cushman M. Identification, synthesis, and biological evaluation of metabolites of the experimental cancer treatment drugs indotecan (LMP400) and indimitecan (LMP776) and investigation of isomerically hydroxylated indenoisoquinoline analogues as topoisomerase I poisons. J Med Chem 2012; 55:10844-62. [PMID: 23215354 PMCID: PMC3542640 DOI: 10.1021/jm300519w] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Hydroxylated analogues of the anticancer topoisomerase I (Top1) inhibitors indotecan (LMP400) and indimitecan (LMP776) have been prepared because (1) a variety of potent Top1 poisons are known that contain strategically placed hydroxyl groups, which provides a clear rationale for incorporating them in the present case, and (2) the hydroxylated compounds could conceivably serve as synthetic standards for the identification of metabolites. Indeed, incubating LMP400 and LMP776 with human liver microsomes resulted in two major metabolites of each drug, which had HPLC retention times and mass fragmentation patterns identical to those of the synthetic standards. The hydroxylated indotecan and indimitecan metabolites and analogues were tested as Top1 poisons and for antiproliferative activity in a variety of human cancer cell cultures and in general were found to be very potent. Differences in activity resulting from the placement of the hydroxyl group are explained by molecular modeling analyses.
Collapse
Affiliation(s)
- Maris A. Cinelli
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy, and the Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, United States
| | - P.V. Narasimha Reddy
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy, and the Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, United States
| | - Peng-Cheng Lv
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy, and the Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, United States
| | - Jian-Hua Liang
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy, and the Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, United States
| | - Lian Chen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The University of Illinois at Chicago, Chicago, IL, 60612, United States
| | - Keli Agama
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-4255, United States
| | - Yves Pommier
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892-4255, United States
| | - Richard B. van Breemen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The University of Illinois at Chicago, Chicago, IL, 60612, United States
| | - Mark Cushman
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy, and the Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, United States
| |
Collapse
|
31
|
Synthesis, characterization, biological studies (DNA binding, cleavage, antibacterial and topoisomerase I) and molecular docking of copper(II) benzimidazole complexes. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2012; 114:15-26. [DOI: 10.1016/j.jphotobiol.2012.05.003] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 04/24/2012] [Accepted: 05/03/2012] [Indexed: 11/19/2022]
|
32
|
Tabassum S, Afzal M, Arjmand F. New heterobimetallic Cu(II)-Sn2(IV) complex as potential topoisomerase I inhibitor: in vitro DNA binding, cleavage and cytotoxicity against human cancer cell lines. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2012; 115:63-72. [PMID: 22884481 DOI: 10.1016/j.jphotobiol.2012.06.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 06/15/2012] [Accepted: 06/26/2012] [Indexed: 11/17/2022]
Abstract
The new heterobimetallic Cu(II)-Sn(2)(IV)/Ni(II)-Sn(2)(IV) complexes 1 and 2 bearing bioactive pharmacophore ligand scaffold; 1,10-phenanthroline and ethylenediamine were synthesized and characterized by spectroscopic (IR, UV-vis, NMR, ESI-MS) and analytical methods. The in vitro DNA binding studies of 1 and 2 with CT-DNA were carried out by employing various biophysical methods which reveal strong electrostatic binding via phosphate backbone of DNA helix, in addition to partial intercalation in the minor groove and stabilized by intramolecular hydrogen bonding. To gain further insight into the molecular recognition at the target site, UV-vis titrations of 1 with 5'-GMP was carried out and validated by (1)H and (31)P NMR. Complex 1 cleaved pBR322 DNA via oxidative pathway and exhibited high inhibition activity against Topo-I at 20 μM. Furthermore, the cytotoxicity of 1 was examined on a panel of human tumor cell lines of different histological origins showing promising antitumor activity.
Collapse
Affiliation(s)
- Sartaj Tabassum
- Department of Chemistry, Aligarh Muslim University, Aligarh 202 002, India.
| | | | | |
Collapse
|
33
|
Arjmand F, Parveen S, Afzal M, Toupet L, Ben Hadda T. Molecular drug design, synthesis and crystal structure determination of CuII–SnIV heterobimetallic core: DNA binding and cleavage studies. Eur J Med Chem 2012; 49:141-50. [DOI: 10.1016/j.ejmech.2012.01.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 01/05/2012] [Accepted: 01/05/2012] [Indexed: 10/14/2022]
|
34
|
Ketron AC, Denny WA, Graves DE, Osheroff N. Amsacrine as a topoisomerase II poison: importance of drug-DNA interactions. Biochemistry 2012; 51:1730-9. [PMID: 22304499 DOI: 10.1021/bi201159b] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Amsacrine (m-AMSA) is an anticancer agent that displays activity against refractory acute leukemias as well as Hodgkin's and non-Hodgkin's lymphomas. The drug is comprised of an intercalative acridine moiety coupled to a 4'-amino-methanesulfon-m-anisidide headgroup. m-AMSA is historically significant in that it was the first drug demonstrated to function as a topoisomerase II poison. Although m-AMSA was designed as a DNA binding agent, the ability to intercalate does not appear to be the sole determinant of drug activity. Therefore, to more fully analyze structure-function relationships and the role of DNA binding in the action of m-AMSA, we analyzed a series of derivatives for the ability to enhance DNA cleavage mediated by human topoisomerase IIα and topoisomerase IIβ and to intercalate DNA. Results indicate that the 3'-methoxy (m-AMSA) positively affects drug function, potentially by restricting the rotation of the headgroup in a favorable orientation. Shifting the methoxy to the 2'-position (o-AMSA), which abrogates drug function, appears to increase the degree of rotational freedom of the headgroup and may impair interactions of the 1'-substituent or other portions of the headgroup within the ternary complex. Finally, the nonintercalative m-AMSA headgroup enhanced enzyme-mediated DNA cleavage when it was detached from the acridine moiety, albeit with 100-fold lower affinity. Taken together, our results suggest that much of the activity and specificity of m-AMSA as a topoisomerase II poison is embodied in the headgroup, while DNA intercalation is used primarily to increase the affinity of m-AMSA for the topoisomerase II-DNA cleavage complex.
Collapse
Affiliation(s)
- Adam C Ketron
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | | | | | | |
Collapse
|
35
|
|
36
|
Bouma MJ, Masson G, Zhu J. Exploiting the Divergent Reactivity of Isocyanoacetates: One-Pot Three-Component Synthesis of Functionalized Angular Furoquinolines. European J Org Chem 2011. [DOI: 10.1002/ejoc.201101567] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
37
|
Kou JF, Qian C, Wang JQ, Chen X, Wang LL, Chao H, Ji LN. Chiral ruthenium(II) anthraquinone complexes as dual inhibitors of topoisomerases I and II. J Biol Inorg Chem 2011; 17:81-96. [PMID: 21858685 DOI: 10.1007/s00775-011-0831-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2011] [Accepted: 07/30/2011] [Indexed: 10/17/2022]
Abstract
DNA topoisomerases (I and II) have been one of the excellent targets in anticancer drug development. Here two chiral ruthenium(II) anthraquinone complexes, Δ- and Λ-[Ru(bpy)(2)(ipad)](2+), where bpy is 2,2'-bipyridine and ipad is 2-(anthracene-9,10-dione-2-yl)imidazo[4,5-f][1,10]phenanthroline, were synthesized and characterized. As expected, both of the Ru(II) complexes intercalate into DNA base pairs and possess an obviously greater affinity with DNA. Topoisomerase inhibition and DNA strand passage assay confirmed that the two complexes are efficient dual inhibitors of topoisomerases I and II by interference with the DNA religation. In MTT cytotoxicity studies, two Ru(II) complexes exhibited antitumor activity against HeLa, MCF-7, HepG2 and BEL-7402 tumor cell lines. Flow cytometry analysis shows an increase in the percentage of cells with apoptotic morphological features in the sub-G1 phase for Ru(II) complexes. Nuclear chromatin cleavage has also been observed from AO/EB staining assay and alkaline single-cell gel electrophoresis (comet assay). The results demonstrated that Δ- and Λ-[Ru(bpy)(2)(ipad)](2+) act as dual inhibitors of topoisomerases I and II, and cause DNA damage that can lead to cell cycle arrest and/or cell death by apoptosis.
Collapse
Affiliation(s)
- Jun-Feng Kou
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
38
|
Synthesis, DNA-binding and topoisomerase inhibitory activity of ruthenium(II) polypyridyl complexes. Eur J Med Chem 2011; 46:1056-65. [DOI: 10.1016/j.ejmech.2011.01.019] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 01/02/2011] [Accepted: 01/12/2011] [Indexed: 11/15/2022]
|
39
|
Gentry AC, Pitts SL, Jablonsky MJ, Bailly C, Graves DE, Osheroff N. Interactions between the etoposide derivative F14512 and human type II topoisomerases: implications for the C4 spermine moiety in promoting enzyme-mediated DNA cleavage. Biochemistry 2011; 50:3240-9. [PMID: 21413765 DOI: 10.1021/bi200094z] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
F14512 is a novel etoposide derivative that contains a spermine in place of the C4 glycosidic moiety. The drug was designed to exploit the polyamine transport system that is upregulated in some cancers. However, a preliminary study suggests that it is also a more efficacious topoisomerase II poison than etoposide [Barret et al. (2008) Cancer Res. 68, 9845-9853]. Therefore, we undertook a more complete study of the actions of F14512 against human type II topoisomerases. As determined by saturation transfer difference (1)H NMR spectroscopy, contacts between F14512 and human topoisomerase IIα in the binary enzyme-drug complex are similar to those of etoposide. Although the spermine of F14512 does not interact with the enzyme, it converts the drug to a DNA binder [Barret et al. (2008)]. Consequently, the influence of the C4 spermine on drug activity was assessed. F14512 is a highly active topoisomerase II poison and stimulates DNA cleavage mediated by human topoisomerase IIα or topoisomerase IIβ. The drug is more potent and efficacious than etoposide or TOP-53, an etoposide derivative that contains a C4 aminoalkyl group that strengthens drug-enzyme binding. Unlike the other drugs, F14512 maintains robust activity in the absence of ATP. The enhanced activity of F14512 correlates with a tighter binding and an increased stability of the ternary topoisomerase II-drug-DNA complex. The spermine-drug core linkage is critical for these attributes. These findings demonstrate the utility of a C4 DNA binding group and provide a rational basis for the development of novel and more active etoposide-based topoisomerase II poisons.
Collapse
Affiliation(s)
- Amanda C Gentry
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-0146, United States
| | | | | | | | | | | |
Collapse
|
40
|
Benzoquinazoline derivatives as new agents affecting DNA processing. Bioorg Med Chem 2011; 19:1197-204. [DOI: 10.1016/j.bmc.2010.12.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 11/29/2010] [Accepted: 12/15/2010] [Indexed: 11/19/2022]
|
41
|
Arjmand F, Muddassir M. Design and synthesis of heterobimetallic topoisomerase I and II inhibitor complexes: in vitro DNA binding, interaction with 5'-GMP and 5'-TMP and cleavage studies. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2010; 101:37-46. [PMID: 20638859 DOI: 10.1016/j.jphotobiol.2010.06.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 06/11/2010] [Accepted: 06/21/2010] [Indexed: 10/19/2022]
Abstract
New potential cancer chemotherapeutic complexes Cu-Sn(2)/Zn-Sn(2) 3 and 4 were designed and prepared as topoisomerases inhibitors; their in vitro DNA binding studies were carried out which reveal strong electrostatic binding via phosphate backbone of DNA helix, in addition to other binding modes viz. coordinate covalent and partial intercalation. To throw insight to molecular binding event at the target site, UV-vis titrations of 3 and 4 with mononucleotides of interest, viz, 5'-GMP and 5'-TMP were carried out, (in case of 4) by (1)H and (31)P NMR. Cleavage studies employing gel electrophoresis demonstrate both the complexes 3 and 4 are efficient cleavage agents and are specific groove binders (complex 3 binds to both major and minor groove while complex 4 is specifically minor groove binder only). In addition, the complexes show high inhibition activity against topoisomerase I and II. However, complex 4 exhibits significant inhibitory effects on the Topo I activity at a very low concentration approximately 2.5 microM.
Collapse
Affiliation(s)
- Farukh Arjmand
- Department of Chemistry, Aligarh Muslim University, Aligarh 202 002, India.
| | | |
Collapse
|
42
|
Novel indoloquinoline derivative, IQDMA, induces G2/M phase arrest and apoptosis in A549 cells through JNK/p38 MAPK signaling activation. Life Sci 2009; 85:505-16. [DOI: 10.1016/j.lfs.2009.08.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 07/27/2009] [Accepted: 08/05/2009] [Indexed: 11/19/2022]
|
43
|
Shuai L, Wang S, Zhang L, Fu B, Zhou X. Cationic Porphyrins and Analogues as New DNA Topoisomerase I and II Inhibitors. Chem Biodivers 2009; 6:827-37. [DOI: 10.1002/cbdv.200800083] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
44
|
Yang SH, Chien CM, Su JC, Chen YL, Chang LS, Lin SR. Novel indoloquinoline derivative, IQDMA, inhibits STAT5 signaling associated with apoptosis in K562 cells. J Biochem Mol Toxicol 2009; 22:396-404. [PMID: 19111001 DOI: 10.1002/jbt.20254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
N'-(11H-indolo[3,2-c]quinolin-6-yl)-N,N-dimethylethane-1,2-diamine (IQDMA), an indoloquinoline derivative, synthesized in our laboratory, has been demonstrated to be an effective antitumor agent in human leukemia cells. In the present study, treatment with IQDMA inhibited phosphorylation of epidermal growth factor receptor (EGFR), Src, Bcr-Abl, and Janus-activated kinase (JAK2) in a time-dependent manner. IQDMA also degraded JAK2 protein. Moreover, signal transducer and activator of transcription 5 (STAT5) signaling were also blocked by IQDMA. However, IQDMA did not inhibit other oncogenic and tumor survival pathways such as those mediated by Akt and extracellular signal-regulated kinase 1/2. Furthermore, IQDMA upregulated the expression of p21 and p27 and downregulated the expression of cyclin D1, myeloid cell leukemia-1(Mcl-1), Bcl-X(L), and vascular endothelial growth factor (VEGF). Taken together, these results indicate that IQDMA causes significant induction of apoptosis in K562 cells via downregulation of EGFR, Src, Bcr-Abl, JAK2, and STAT5 signaling and modulation of p21, p27, cyclin D1, Mcl-1, Bcl-X(L), and VEGF proteins. Thus, IQDMA appears to be a potential therapeutic agent for treating leukemia K562 cells.
Collapse
Affiliation(s)
- Sheng-Huei Yang
- Faculty of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
45
|
Kouznetsov VV. Recent synthetic developments in a powerful imino Diels–Alder reaction (Povarov reaction): application to the synthesis of N-polyheterocycles and related alkaloids. Tetrahedron 2009. [DOI: 10.1016/j.tet.2008.12.059] [Citation(s) in RCA: 455] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
46
|
Tomoda K, Ohkoshi T, Hirota K, Sonavane GS, Nakajima T, Terada H, Komuro M, Kitazato K, Makino K. Preparation and properties of inhalable nanocomposite particles for treatment of lung cancer. Colloids Surf B Biointerfaces 2009; 71:177-82. [PMID: 19264458 DOI: 10.1016/j.colsurfb.2009.02.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Revised: 02/02/2009] [Accepted: 02/04/2009] [Indexed: 11/28/2022]
Abstract
Nanoparticles have widely been studied in drug delivery research for targeting and controlled release. The aim of this article is application of nanoparticles as an inhalable agent for treatment of lung cancer. To deposit effectively deep the particles in the lungs, the PLGA nanoparticles loaded with the anticancer drug 6-{[2-(dimethylamino)ethyl]amino}-3-hydroxyl-7H-indeno[2,1-c]quinolin-7-one dihydrochloride (TAS-103) were prepared in the form of nanocomposite particles. The nanocomposite particles consist of the complex of drug-loaded nanoparticles and excipients. In this study, the anticancer effects of the nanocomposite particles against the lung cancer cell line A549. Also, the concentration of TAS-103 in blood and lungs were determined after administration of the nanocomposite particles by inhalation to rats. TAS-103-loaded PLGA nanoparticles were prepared with 5% and 10% of loading ratio by spray drying method with trehalose as an excipient. The 5% drug-loaded nanocomposite particles were more suitable for inhalable agent because of the sustained release of TAS-103 and higher FPF value. Cytotoxicity of nanocomposite particles against A549 cells was higher than that of free drug. When the nanocomposite particles were administered in rats by inhalation, drug concentration in lung was much higher than that in plasma. Furthermore, drug concentration in lungs administered by inhalation of nanocomposite particles was much higher than that after intravenous administration of free drug. From these results, the nanocomposite particle systems could be promising for treatment of lung cancer.
Collapse
Affiliation(s)
- Keishiro Tomoda
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Oppegard LM, Ougolkov AV, Luchini DN, Schoon RA, Goodell JR, Kaur H, Billadeau DD, Ferguson DM, Hiasa H. Novel acridine-based compounds that exhibit an anti-pancreatic cancer activity are catalytic inhibitors of human topoisomerase II. Eur J Pharmacol 2008; 602:223-9. [PMID: 19071108 DOI: 10.1016/j.ejphar.2008.11.044] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 11/03/2008] [Accepted: 11/18/2008] [Indexed: 10/21/2022]
Abstract
We have identified a small library of novel substituted 9-aminoacridine derivatives that inhibit cell proliferation of pancreatic cancer cell lines by inducing apoptosis [Goodell, J.R. et al., 2008. J. Med. Chem. 51, 179-182.]. To further investigate their antiproliferative activities, we have assessed the antiproliferative activity of these acridine-based compounds against several pancreatic cancer cell lines. All four compounds used in this study inhibited the proliferation of pancreatic cancer cell lines in vitro. In addition, we have employed a xenograft tumor model and found that these compounds also inhibit the proliferation of pancreatic cancer in vivo. In light of the potential importance of the anticancer activity of these acridine-based compounds, we have conducted a series of biochemical assays to determine the effect of these compounds on human topoisomerase II. Unlike amsacrine, these compounds do not poison topoisomerase II. Similar to amsacrine, however, these compounds intercalate into DNA in a way that they would alter the apparent topology of the DNA substrate. Thus, inhibition of the relaxation activity of topoisomerase II by these compounds has been reexamined using a DNA strand passage assay. We have found that these compounds, indeed, inhibit the catalytic activity of topoisomerase II. Thus, these novel acridine-based compounds with anti-pancreatic cancer activity are catalytic inhibitors, not poisons, of human topoisomerase II.
Collapse
Affiliation(s)
- Lisa M Oppegard
- Department of Pharmacology, University of Minnesota Medical School-Twin Cities, Minneapolis, Minnesota 55455, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Gao F, Chao H, Ji LN. DNA Binding, Photocleavage, and Topoisomerase Inhibition of Functionalized Ruthenium(II)-Polypyridine Complexes. Chem Biodivers 2008; 5:1962-1979. [DOI: 10.1002/cbdv.200890181] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
49
|
Jobson AG, Willmore E, Tilby MJ, Mistry P, Charlton P, Austin CA. Effect of phenazine compounds XR11576 and XR5944 on DNA topoisomerases. Cancer Chemother Pharmacol 2008; 63:889-901. [PMID: 18679685 DOI: 10.1007/s00280-008-0812-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Accepted: 07/20/2008] [Indexed: 11/26/2022]
Abstract
PURPOSE Previous in vitro cleavage data showed that XR11576 and XR5944 stabilised topoisomerase I and topoisomerase II complexes on DNA in a dose-dependent fashion. However, some studies indicated a possible topoisomerase-independent mechanism of action for these drugs. METHODS Three methods, the TARDIS assay, immunoband depletion and the K(+)/SDS assay have been used to assess topoisomerase complex formation induced by XR11576 or XR5944 in human leukaemic K562 cells. RESULTS TARDIS and immunoband depletion assays demonstrated that XR11576 and XR5944 induced complex formation for both topoisomerase I and topoisomerase II (alpha and beta) in a dose- and time-dependent manner, following exposure times of 24 and 48 h at concentrations of 1 or 10 microM. The K(+)/SDS assay showed the formation of protein/DNA complexes after a 1 h exposure to 1 or 10 muM XR11576. CONCLUSION Our data confirm that XR11576 or XR5944 can form topoisomerase complexes, after long periods of exposure.
Collapse
Affiliation(s)
- Andrew G Jobson
- Institute for Cell and Molecular Biosciences, The Medical School, Newcastle University, Newcastle Upon Tyne, NE2 4HH, UK
| | | | | | | | | | | |
Collapse
|
50
|
Chien CM, Yang SH, Lin KL, Chen YL, Chang LS, Lin SR. Novel indoloquinoline derivative, IQDMA, suppresses STAT5 phosphorylation and induces apoptosis in HL-60 cells. Chem Biol Interact 2008; 176:40-7. [PMID: 18638462 DOI: 10.1016/j.cbi.2008.06.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Revised: 06/16/2008] [Accepted: 06/18/2008] [Indexed: 12/16/2022]
Abstract
Signal transducers and activators of transcription (STATs) are a family proteins that mediate cytokine and growth factor-induced signals playing a role in cell differentiation, proliferation, angiogenesis, and apoptosis. One STAT family member, STAT5, is often constitutively active in myeloid leukaemia. Agents that can suppress STAT5 activation have potential for prevention and treatment of cancer. N'-(11H-indolo[3,2-c]quinolin-6-yl)-N,N-dimethylethane-1,2-dia-mine (IQDMA), an indoloquinoline derivative, synthesized in our laboratory, has been demonstrated to be an effective anti-tumor agent in human leukemia cells. In the present report, we tested IQDMA for its ability to suppress STAT5 activation. We found that IQDMA inhibited constitutive activation of STAT5 in HL-60 cells in a dose- and time-dependent manner. The activation of Src and interleukin-6 (IL-6), implicated in STAT5 activation, was also inhibited by the IQDMA. Furthermore, IQDMA up-regulated Bax, and down-regulated Bcl-2, Bcl-X(L), cyclin D1, and vascular endothelial growth factor (VEGF) as followed by growth arrest of HL-60 cells, but the expression of survivin did not change in the presence of IQDMA. Taken together, these results indicate that IQDMA causes significant induction of apoptosis in HL-60 cells via down-regulation of Src, IL-6, and STAT5 signaling and modulation of Bcl-2 family, cyclin D1 and VEGF proteins. Thus, IQDMA appears to be a potential therapeutic agent for treating leukaemia HL-60 cells.
Collapse
Affiliation(s)
- Ching-Ming Chien
- Faculty of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|