1
|
Khaled DM, Elshakre ME, Noamaan MA, Butt H, Abdel Fattah MM, Gaber DA. A Computational QSAR, Molecular Docking and In Vitro Cytotoxicity Study of Novel Thiouracil-Based Drugs with Anticancer Activity against Human-DNA Topoisomerase II. Int J Mol Sci 2022; 23:11799. [PMID: 36233102 PMCID: PMC9570267 DOI: 10.3390/ijms231911799] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 11/30/2022] Open
Abstract
Computational chemistry, molecular docking, and drug design approaches, combined with the biochemical evaluation of the antitumor activity of selected derivatives of the thiouracil-based dihydroindeno pyrido pyrimidines against topoisomerase I and II. The IC50 of other cell lines including the normal human lung cell line W138, lung cancer cell line, A549, breast cancer cell line, MCF-7, cervical cancer, HeLa, and liver cancer cell line HepG2 was evaluated using biochemical methods. The global reactivity descriptors and physicochemical parameters were computed, showing good agreement with the Lipinski and Veber's rules of the drug criteria. The molecular docking study of the ligands with the topoisomerase protein provides the binding sites, binding energies, and deactivation constant for the inhibition pocket. Various biochemical methods were used to evaluate the IC50 of the cell lines. The QSAR model was developed for colorectal cell line HCT as a case study. Four QSAR statistical models were predicted between the IC50 of the colorectal cell line HCT to correlate the anticancer activity and the computed physicochemical and quantum chemical global reactivity descriptors. The predictive power of the models indicates a good correlation between the observed and the predicted activity.
Collapse
Affiliation(s)
- Doaa M. Khaled
- Histology and Cytology Department, Faculty of Medicine, Helwan University, Cairo 11795, Egypt
| | - Mohamed E. Elshakre
- Chemistry Department, Faculty of Science, Cairo University, Cairo 12613, Egypt
| | - Mahmoud A. Noamaan
- Mathematics Department, Faculty of Science, Cairo University, Cairo 12613, Egypt
| | - Haider Butt
- Department of Mechanical Engineering, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Marwa M. Abdel Fattah
- Histology and Cytology Department, Faculty of Medicine, Misr University for Science & Technology, Cairo P.O. Box 77, Egypt
| | - Dalia A. Gaber
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Helwan University, Cairo 11795, Egypt
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates
| |
Collapse
|
2
|
Xiao L, Xu J, Weng Q, Zhou L, Wang M, Liu M, Li Q. Mechanism of a Novel Camptothecin-Deoxycholic Acid Derivate Induced Apoptosis against Human Liver Cancer HepG2 Cells and Human Colon Cancer HCT116 Cells. Recent Pat Anticancer Drug Discov 2019; 14:370-382. [PMID: 31644410 DOI: 10.2174/1574892814666191016162346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 09/25/2019] [Accepted: 10/09/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Camptothecin (CPT) is known as an anticancer drug in traditional Chinese medicine. However, due to the lack of targeting, low solubility, and instability of CPT, its therapeutic applications are hampered. Therefore, we synthesized a series of CPT-bile acid analogues that obtained a national patent to improve their tumour-targeting chemotherapeutic effects on liver or colon cancers. Among these analogues, the compound G2 shows high antitumor activity with enhanced liver targeting and improved oral absorption. It is significant to further investigate the possible anticancer mechanism of G2 for its further clinical research and application. OBJECTIVE We aimed to unearth the anticancer mechanism of G2 in HepG2 and HCT116 cells. METHODS Cell viability was measured using MTT assay; cell cycle, Mitochondrial Membrane Potential (MMP), and cell apoptosis were detected by flow cytometer; ROS was measured by Fluorescent Microplate Reader; the mRNA and protein levels of cell cycle-related and apoptosis-associated proteins were examined by RT-PCR and western blot, respectively. RESULTS We found that G2 inhibited cells proliferation of HepG2 and HCT116 remarkably in a dosedependent manner. Moreover, G2-treatment led to S and G2/M phase arrest in both cells, which could be elucidated by the change of mRNA levels of p21, p27 and Cyclin E and the increased protein level of p21. G2 also induced dramatically ROS accumulated and MMP decreased, which contributed to the apoptosis through activation of both the extrinsic and intrinsic pathways via changing the genes and proteins expression involved in apoptosis pathway in both of HepG2 and HCT116 cells. CONCLUSION These findings suggested that the apoptosis in both cell lines induced by G2 was related to the extrinsic and intrinsic pathways.
Collapse
Affiliation(s)
- Linxia Xiao
- Collaborative Innovation Center of Yangtze River Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Jialin Xu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Qi Weng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Leilei Zhou
- Collaborative Innovation Center of Yangtze River Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Mengke Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Miao Liu
- College of Life Sciences, Northeast Agricultural University, Harbin, China
| | - Qingyong Li
- Collaborative Innovation Center of Yangtze River Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China.,College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
3
|
Mechanisms underlying synergy between DNA topoisomerase I-targeted drugs and mTOR kinase inhibitors in NF1-associated malignant peripheral nerve sheath tumors. Oncogene 2019; 38:6585-6598. [PMID: 31444410 DOI: 10.1038/s41388-019-0965-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/08/2019] [Accepted: 05/27/2019] [Indexed: 01/08/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are soft-tissue sarcomas that frequently arise in patients with neurofibromatosis type 1 (NF1). Most of these tumors are unresectable at diagnosis and minimally responsive to conventional treatment, lending urgency to the identification of new pathway dependencies and drugs with potent antitumor activities. We therefore examined a series of candidate agents for their ability to induce apoptosis in MPNST cells arising in nf1/tp53-deficient zebrafish. In this study, we found that DNA topoisomerase I-targeted drugs and mTOR kinase inhibitors were the most effective single agents in eliminating MPNST cells without prohibitive toxicity. In addition, three members of these classes of drugs, either AZD2014 or INK128 in combination with irinotecan, acted synergistically to induce apoptosis both in vitro and in vivo. In mechanistic studies, irinotecan not only induces apoptosis by eliciting a DNA damage response, but also acts synergistically with AZD2014 to potentiate the hypophosphorylation of 4E-BP1, a downstream target of mTORC1. Profound hypophosphorylation of 4E-BP1 induced by this drug combination causes an arrest of protein synthesis, which potently induces tumor cell apoptosis. Our findings provide a compelling rationale for further in vivo evaluation of the combination of DNA topoisomerase I-targeted drugs and mTOR kinase inhibitors against these aggressive nerve sheath tumors.
Collapse
|
4
|
Cinelli MA. Topoisomerase 1B poisons: Over a half-century of drug leads, clinical candidates, and serendipitous discoveries. Med Res Rev 2018; 39:1294-1337. [PMID: 30456874 DOI: 10.1002/med.21546] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 12/17/2022]
Abstract
Topoisomerases are DNA processing enzymes that relieve supercoiling (torsional strain) in DNA, are necessary for normal cellular division, and act by nicking (and then religating) DNA strands. Type 1B topoisomerase (Top1) is overexpressed in certain tumors, and the enzyme has been extensively investigated as a target for cancer chemotherapy. Various chemical agents can act as "poisons" of the enzyme's religation step, leading to Top1-DNA lesions, DNA breakage, and eventual cellular death. In this review, agents that poison Top1 (and have thus been investigated for their anticancer properties) are surveyed, including natural products (such as camptothecins and indolocarbazoles), semisynthetic camptothecin and luotonin derivatives, and synthetic compounds (such as benzonaphthyridines, aromathecins, and indenoisoquinolines), as well as targeted therapies and conjugates. Top1 has also been investigated as a therapeutic target in certain viral and parasitic infections, as well as autoimmune, inflammatory, and neurological disorders, and a summary of literature describing alternative indications is also provided. This review should provide both a reference for the medicinal chemist and potentially offer clues to aid in the development of new Top1 poisons.
Collapse
Affiliation(s)
- Maris A Cinelli
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
5
|
Dai Y, Hua Q, Ling J, Shao C, Zhong C, Zhang X, Hu Y, Zhang L, Liu Y. Quantum chemical calculation of free radical substitution reaction mechanism of camptothecin. J Mol Graph Model 2018; 84:174-181. [PMID: 30015049 DOI: 10.1016/j.jmgm.2018.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/31/2022]
Abstract
Free radical substitution reaction, which has low energy barrier and takes place in mild reaction conditions, is an important method for camptothecin's modification. The experimental data show that the free radical substitution reaction of camptothecin has high site selectivity, and prefers to take place at site 7. Up to now, few researches focus on the mechanism of it. In this study, the differences of the reaction rate constant (k) for the reactions at different sites, such as site of 7, 9, 10, 11, 12, were investigated with B3LYP of density functional theory at the 6-31 + G (d, p) base set level and CPCM aqueous solvent model. It was found that the substitution reaction can be carried out in two steps in acidic condition. First, the methyl radical attacks the corresponding site to form an intermediate having methyl radical combined with the camptothecin skeleton, and then a hydrogen atom was abstracted by the singlet oxygen to form methyl camptothecin, wherein the first step was the rate control step of the reaction. The results show that site 7 has the higherreaction rate constant (k) than other examined sites, indicating that the reaction tends to take place on site 7 position, which is in agreement with the experimental results.
Collapse
Affiliation(s)
- Yujie Dai
- Key Laboratory of Industrial Fermentation Microbiology (Tianjin University of Science & Technology), Ministry of Education, College of Bioengineering, Tianjin University of Science and Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, PR China.
| | - Qingyuan Hua
- Key Laboratory of Industrial Fermentation Microbiology (Tianjin University of Science & Technology), Ministry of Education, College of Bioengineering, Tianjin University of Science and Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, PR China
| | - Jun Ling
- Key Laboratory of Industrial Fermentation Microbiology (Tianjin University of Science & Technology), Ministry of Education, College of Bioengineering, Tianjin University of Science and Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, PR China
| | - Chunfu Shao
- Key Laboratory of Industrial Fermentation Microbiology (Tianjin University of Science & Technology), Ministry of Education, College of Bioengineering, Tianjin University of Science and Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, PR China
| | - Cheng Zhong
- Key Laboratory of Industrial Fermentation Microbiology (Tianjin University of Science & Technology), Ministry of Education, College of Bioengineering, Tianjin University of Science and Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, PR China
| | - Xiuli Zhang
- Key Laboratory of Industrial Fermentation Microbiology (Tianjin University of Science & Technology), Ministry of Education, College of Bioengineering, Tianjin University of Science and Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, PR China
| | - Yanying Hu
- Key Laboratory of Industrial Fermentation Microbiology (Tianjin University of Science & Technology), Ministry of Education, College of Bioengineering, Tianjin University of Science and Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, PR China
| | - Liming Zhang
- Key Laboratory of Industrial Fermentation Microbiology (Tianjin University of Science & Technology), Ministry of Education, College of Bioengineering, Tianjin University of Science and Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, PR China
| | - Yaotian Liu
- Key Laboratory of Industrial Fermentation Microbiology (Tianjin University of Science & Technology), Ministry of Education, College of Bioengineering, Tianjin University of Science and Technology, No.29 of 13th Street, TEDA, Tianjin, 300457, PR China
| |
Collapse
|
6
|
Laev SS, Salakhutdinov NF, Lavrik OI. Tyrosyl-DNA phosphodiesterase inhibitors: Progress and potential. Bioorg Med Chem 2016; 24:5017-5027. [PMID: 27687971 DOI: 10.1016/j.bmc.2016.09.045] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/30/2016] [Accepted: 09/18/2016] [Indexed: 10/21/2022]
Abstract
DNA topoisomerases are essential during transcription and replication. The therapeutic mechanism of action of topoisomerase inhibitors is enzyme poisoning rather than catalytic inhibition. Tyrosyl-DNA phosphodiesterases 1 or 2 were found as DNA repair enzymes hydrolyzing the covalent bond between the tyrosyl residue of topoisomerases I or II and the 3'- or 5'-phosphate groups in DNA, respectively. Tyrosyl-DNA phosphodiesterase 1 is a key enzyme in DNA repair machinery and a promising target for antitumor and neurodegenerative therapy. Inhibitors of tyrosyl-DNA phosphodiesterase 1 could act synergistically with topoisomerase I inhibitors and thereby potentiate the effects of topoisomerase I poisons. Tyrosyl-DNA phosphodiesterase 2 is an enzyme that specifically repairs DNA damages induced by topoisomerase II poisons and causes resistance to these drugs. Selective inhibition of tyrosyl-DNA phosphodiesterase 2 may be a novel approach to overcome intrinsic or acquired resistance to topoisomerase II-targeted drug therapy. Thus, agents that inhibit tyrosyl-DNA phosphodiesterases 1 and 2 have many applications in biochemical and physiological research and they have the potential to become anticancer and antiviral drugs. The structures, mechanism of action and therapeutic rationale of tyrosyl-DNA phosphodiesterase inhibitors and their development for combinations with topoisomerase inhibitors and DNA damaging agents are discussed.
Collapse
Affiliation(s)
- Sergey S Laev
- Vorozhtsov Institute of Organic Chemistry, Siberian Division, Russian Academy of Sciences, pr. akademika Lavrent'eva 9, Novosibirsk 630090, Russian Federation.
| | - Nariman F Salakhutdinov
- Vorozhtsov Institute of Organic Chemistry, Siberian Division, Russian Academy of Sciences, pr. akademika Lavrent'eva 9, Novosibirsk 630090, Russian Federation; Novosibirsk State University, Pirogova Str. 2, Novosibirsk 630090, Russian Federation
| | - Olga I Lavrik
- Novosibirsk State University, Pirogova Str. 2, Novosibirsk 630090, Russian Federation; Novosibirsk Institute of Chemical Biology and Fundamental Medicine, Siberian Division, Russian Academy of Sciences, pr. akademika Lavrent'eva 8, Novosibirsk 630090, Russian Federation
| |
Collapse
|
7
|
Wu D, Shi W, Zhao J, Wei Z, Chen Z, Zhao D, Lan S, Tai J, Zhong B, Yu H. Assessment of the chemotherapeutic potential of a new camptothecin derivative, ZBH-1205. Arch Biochem Biophys 2016; 604:74-85. [PMID: 27302903 DOI: 10.1016/j.abb.2016.06.007] [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: 04/12/2016] [Revised: 05/26/2016] [Accepted: 06/09/2016] [Indexed: 10/21/2022]
Abstract
CPT-11 (irinotecan) is a derivative of camptothecin which is a natural product derived from the Chinese tree Camptotheca acuminta and widely used in antitumor therapy. Here, the in vitro anti-tumor activity and associated mechanisms of a novel derivative of camptothecin, ZBH-1205, were investigated in a panel of 9 human tumor cell lines, as well as in HEK 293 and SK-OV-3/DPP, a multi-drug resistant (MDR) cell line, and compared to CPT-11 and 7-ethyl-10-hydroxy-camptothecin (SN38). Comparisons between the different compounds were made on the basis of IC50 values as determined by the MTT assay, and flow cytometry was used to evaluate cell cycle progression, apoptosis, and the levels of pro- and active caspase-3 among different treatment groups. Interaction between the molecules and topoisomerase-1 (Topo-1)-DNA complexes was detected by a DNA relaxation assay. Our results demonstrated that IC50 values for ZBH-1205 ranged from 0.0009 μmol/L to 2.5671 μmol/L, which were consistently lower than IC50 values of CPT-11 or SN38 in the panel of cell lines, including SK-OV-3/DPP. Furthermore, ZBH-1205 was more effective than CPT-11 or SN38 at stabilizing Topo-1-DNA complexes and inducing tumor cell apoptosis. Therefore, ZBH-1205 is a promising chemotherapeutic agent to be further assessed in large-scale clinical trials.
Collapse
Affiliation(s)
- Di Wu
- Tumor Center of Jilin University No.1 Hospital, Changchun 130021, China
| | - Weiguo Shi
- Institute of Pharmacology and Toxicology Academy of Military Medical Sciences, Beijing 100850, China
| | - Jing Zhao
- Key Lab for Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun 130062, China
| | - Zhengren Wei
- Department of Pharmacology, Bethune Medical College, Jilin University, Changchun 130021, China
| | - Zhijia Chen
- Department of Pharmacology, Bethune Medical College, Jilin University, Changchun 130021, China
| | - Dawei Zhao
- Jilin Province Tumor Hospital, Changchun 130021, China
| | - Shijie Lan
- Tumor Center of Jilin University No.1 Hospital, Changchun 130021, China
| | - Jiandong Tai
- Tumor Center of Jilin University No.1 Hospital, Changchun 130021, China.
| | - Bohua Zhong
- Institute of Pharmacology and Toxicology Academy of Military Medical Sciences, Beijing 100850, China.
| | - Hong Yu
- Cell Biology Laboratory, Jilin Province Tumor Institute, Changchun 130021, China.
| |
Collapse
|
8
|
Casadó A, Giuffrida MC, Sagristá ML, Castelli F, Pujol M, Alsina MA, Mora M. Langmuir monolayers and Differential Scanning Calorimetry for the study of the interactions between camptothecin drugs and biomembrane models. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:422-33. [DOI: 10.1016/j.bbamem.2015.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/20/2015] [Accepted: 12/03/2015] [Indexed: 10/22/2022]
|
9
|
Li Q, Liu T, Li Y, Luo S, Zhu Q, Zhang L, Zhao T. Transport and killing mechanism of a novel camptothecin-deoxycholic acid derivate on hepatocellular carcinoma cells. J Drug Target 2014; 22:543-52. [PMID: 24725118 DOI: 10.3109/1061186x.2014.906603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Abstract Camptothecin-20(s)-O-glycine ester-[N-(3'α, 12'α-dihydroxy-24'-carbonyl-5'β-cholan)] (A2), 10-(3'α,12'α-dihydroxy-5'β-cholan-24'-carboxyl)-(20 s)-camptothecin (C2), and 10-O-(3-O-(3'α, 12'α-dihydroxy-24'-carbonyl-5'β-cholan)-propyl)-(20S)-camptothecin (D2) are novel camptothecin-deoxycholic acid analogues. MTT assays were performed to assess the anticancer activity of these compounds against hepatocellular carcinoma SMMC-7721, breast carcinoma MCF-7, and colorectal carcinoma HCT-116 cells. A2 had a high killing ability on SMMC-7721 cells selectively, but C2 and D2 did not exhibit selectivity with regard to SMMC-7721 killing. Uptake assays were performed in an effort to elucidate the transport mechanisms of A2 into SMMC-7721 cells. A2 increased the mRNA expression of OATP1B3 (an organic anion-transporting polypeptide) and uptake of A2 was inhibited by rifampin (inhibitor of OATP1B3), which indicated that the transporter-mediated transport of A2 was mediated by OATP1B3. In addition, according to the western blot and apoptosis assays, we found that A2 killed SMMC-7721 cells by inducing cell apoptosis mainly via an AIF (apoptosis-inducing factor) pathway and a caspase-dependent mitochondria apoptosis pathway.
Collapse
Affiliation(s)
- Qingyong Li
- College of Pharmaceutical Science, Zhejiang University of Technology , Hangzhou , China
| | | | | | | | | | | | | |
Collapse
|
10
|
|
11
|
Acevedo-Morantes CY, Acevedo-Morantes MT, Suleiman-Rosado D, Ramírez-Vick JE. Evaluation of the cytotoxic effect of camptothecin solid lipid nanoparticles on MCF7 cells. Drug Deliv 2013; 20:338-48. [PMID: 24024505 DOI: 10.3109/10717544.2013.834412] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Camptothecin (CPT) and its analogs exhibit remarkable anti-tumor activity, due to their ability to inhibit DNA topoisomerase I. However, its use is limited by the lack of solubility and stability of the active lactone form. An attractive alternative is the encapsulation of CPT within liposomes. In this study, CPT was incorporated into solid lipid nanoparticles (SLN) based on the triglyceride, Compritol 888 ATO, using supercritical fluid technology without requiring the use of harmful solvents. This drug delivery system was characterized and its cytotoxicity effect was evaluated by measuring MCF7 and MCF10A cell viability as a function of drug loading during a 48-h treatment. Results showed that after 10 h of treatment, MCF7 cells displayed an IC50 of 0.23±0.034 μM at a 1:5 (CPT:SLN) loading and 0.22±0.027 μM at a 1:10 loading, whereas MCF10A cells displayed an IC50 of 0.40±0.036 μM at 1:5 and 0.60±0.063 μM at 1:10. On the other hand, the IC50 of free CPT was 0.57±0.035 μM and 1.07±0.077 μM for MCF7 and MCF10A cells, respectively. Cellular uptake and retention measurements in both cells displayed a two-fold increase when using the SLN formulation. The results from this study showed that the cytotoxic effects of CPT in a SLN formulation improved when compared with those seen with free CPT. The results of this study showed that delivery of CPT as a SLN formulation could be a promising strategy for enhancing its chemotherapeutic effects.
Collapse
|
12
|
Huang Q, Wang L, Lu W. Evolution in medicinal chemistry of E-ring-modified Camptothecin analogs as anticancer agents. Eur J Med Chem 2013; 63:746-57. [DOI: 10.1016/j.ejmech.2013.01.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 01/13/2013] [Accepted: 01/16/2013] [Indexed: 12/24/2022]
|
13
|
Total synthesis of a novel non-lactone camptothecin analog through microwave-assisted [3,3]-sigmatropic rearrangement. Tetrahedron 2013. [DOI: 10.1016/j.tet.2013.01.091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
14
|
Huehls AM, Wagner JM, Huntoon CJ, Karnitz LM. Identification of DNA repair pathways that affect the survival of ovarian cancer cells treated with a poly(ADP-ribose) polymerase inhibitor in a novel drug combination. Mol Pharmacol 2012; 82:767-76. [PMID: 22833573 PMCID: PMC3463227 DOI: 10.1124/mol.112.080614] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 07/24/2012] [Indexed: 12/17/2022] Open
Abstract
Floxuridine (5-fluorodeoxyuridine, FdUrd), a U.S. Food and Drug Administration-approved drug and metabolite of 5-fluorouracil, causes DNA damage that is repaired by base excision repair (BER). Thus, poly(ADP-ribose) polymerase (PARP) inhibitors, which disrupt BER, markedly sensitize ovarian cancer cells to FdUrd, suggesting that this combination may have activity in this disease. It remains unclear, however, which DNA repair and checkpoint signaling pathways affect killing by these agents individually and in combination. Here we show that depleting ATR, BRCA1, BRCA2, or RAD51 sensitized to ABT-888 (veliparib) alone, FdUrd alone, and FdUrd + ABT-888 (F+A), suggesting that homologous recombination (HR) repair protects cells exposed to these agents. In contrast, disabling the mismatch, nucleotide excision, Fanconi anemia, nonhomologous end joining, or translesion synthesis repair pathways did not sensitize to these agents alone (including ABT-888) or in combination. Further studies demonstrated that in BRCA1-depleted cells, F+A was more effective than other chemotherapy+ABT-888 combinations. Taken together, these studies 1) identify DNA repair and checkpoint pathways that are important in ovarian cancer cells treated with FdUrd, ABT-888, and F+A, 2) show that disabling HR at the level of ATR, BRCA1, BRCA2, or RAD51, but not Chk1, ATM, PTEN, or FANCD2, sensitizes cells to ABT-888, and 3) demonstrate that even though ABT-888 sensitizes ovarian tumor cells with functional HR to FdUrd, the effects of this drug combination are more profound in tumors with HR defects, even compared with other chemotherapy + ABT-888 combinations, including cisplatin + ABT-888.
Collapse
Affiliation(s)
- Amelia M Huehls
- Division of Oncology Research, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, College of Medicine, Rochester, Minnesota, USA
| | | | | | | |
Collapse
|
15
|
Ye D, Shi Q, Leung CH, Kim SW, Park SY, Gullen EA, Jiang ZL, Zhu H, Morris-Natschke SL, Cheng YC, Lee KH. Antitumor agents 294. Novel E-ring-modified camptothecin-4β-anilino-4'-O-demethyl-epipodophyllotoxin conjugates as DNA topoisomerase I inhibitors and cytotoxic agents. Bioorg Med Chem 2012; 20:4489-94. [PMID: 22698783 DOI: 10.1016/j.bmc.2012.05.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 05/04/2012] [Accepted: 05/12/2012] [Indexed: 10/28/2022]
Abstract
Two conjugates (1 and 2) of camptothecin (CPT) and 4β-anilino-4'-O-demethylepipodophyllotoxin were previously shown to exert antitumor activity through inhibition of topoisomerase I (topo I). In this current study, two novel conjugates (1E and 2E) with an open E-ring in the CPT moiety were first synthesized and evaluated for biological activity in comparison with their intact E-ring congeners. This novel class of CPT-derivatives exhibits its antitumor effect against CPT-sensitive and -resistant cells, in part, by inhibiting topo I-linked DNA (TLD) religation. An intact E-ring was not essential for the inhibition of TLD religation, although conjugates with an open E-ring were less potent than the closed ring analogs. This lower religation potency resulted in decreased formation of protein-linked DNA breaks (PLDBs), and hence, less cell growth inhibition. In addition to their impact on topo I, conjugates 1E, 2, and 2E exhibited a minor inhibitory effect on topo II-induced DNA cleavage. The novel structures of 1E and 2E may present scaffolds for further development of dual function topo I and II inhibitors with improved pharmacological profiles and physicochemical properties.
Collapse
Affiliation(s)
- Deyong Ye
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7568, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Exploring DNA topoisomerase I ligand space in search of novel anticancer agents. PLoS One 2011; 6:e25150. [PMID: 21966440 PMCID: PMC3178613 DOI: 10.1371/journal.pone.0025150] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 08/29/2011] [Indexed: 01/22/2023] Open
Abstract
DNA topoisomerase I (Top1) is over-expressed in tumour cells and is an important target in cancer chemotherapy. It relaxes DNA torsional strain generated during DNA processing by introducing transient single-strand breaks and allowing the broken strand to rotate around the intermediate Top1 – DNA covalent complex. This complex can be trapped by a group of anticancer agents interacting with the DNA bases and the enzyme at the cleavage site, preventing further topoisomerase activity. Here we have identified novel Top1 inhibitors as potential anticancer agents by using a combination of structure- and ligand-based molecular modelling methods. Pharmacophore models have been developed based on the molecular characteristics of derivatives of the alkaloid camptothecin (CPT), which represent potent antitumour agents and the main group of Top1 inhibitors. The models generated were used for in silico screening of the National Cancer Institute (NCI, USA) compound database, leading to the identification of a set of structurally diverse molecules. The strategy is validated by the observation that amongst these molecules are several known Top1 inhibitors and agents cytotoxic against human tumour cell lines. The potential of the untested hits to inhibit Top1 activity was further evaluated by docking into the binding site of a Top1 – DNA complex, resulting in a selection of 10 compounds for biological testing. Limited by the compound availability, 7 compounds have been tested in vitro for their Top1 inhibitory activity, 5 of which display mild to moderate Top1 inhibition. A further compound, found by similarity search to the active compounds, also shows mild activity. Although the tested compounds display only low in vitro antitumour activity, our approach has been successful in the identification of structurally novel Top1 inhibitors worthy of further investigation as potential anticancer agents.
Collapse
|
17
|
Evdokimov NM, Van slambrouck S, Heffeter P, Tu L, Le Calvé B, Lamoral-Theys D, Hooten CJ, Uglinskii PY, Rogelj S, Kiss R, Steelant WF, Berger W, Yang JJ, Bologa CG, Kornienko A, Magedov IV. Structural simplification of bioactive natural products with multicomponent synthesis. 3. Fused uracil-containing heterocycles as novel topoisomerase-targeting agents. J Med Chem 2011; 54:2012-21. [PMID: 21388138 PMCID: PMC3087497 DOI: 10.1021/jm1009428] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
After the initial discovery of antiproliferative and apoptosis-inducing properties of a camptothecin-inspired pentacycle based on a 1H-indeno[2',1':5,6]dihydropyrido[2,3-d]pyrimidine scaffold, a library of its analogues as well as their oxidized planar counterparts were prepared utilizing a practical multicomponent synthetic protocol. The synthesized compounds exhibited submicromolar to low micromolar antiproliferative potencies toward a panel of human cancer cell lines. Biochemical experiments are consistent with the dihydropyridine library members undergoing intracellular oxidation to the corresponding planar pyridines, which then inhibit topoisomerase II activity, leading to inhibition of proliferation and cell death. Because of facile synthetic preparation and promising antitopoisomerase activity, both the dihydropyridine and planar pyridine-based compounds represent a convenient starting point for anticancer drug discovery.
Collapse
Affiliation(s)
- Nikolai M. Evdokimov
- Department of Chemistry, New Mexico Institute of Mining and Technology, Socorro, New Mexico 87801, USA
| | - Severine Van slambrouck
- Department of Chemistry, New Mexico Institute of Mining and Technology, Socorro, New Mexico 87801, USA
| | - Petra Heffeter
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| | - Lee Tu
- Department of Chemistry, New Mexico Institute of Mining and Technology, Socorro, New Mexico 87801, USA
| | - Benjamin Le Calvé
- Laboratoire de Toxicologie and Laboratoire de Chimie Analytique, Toxicologie et Chimie Physique Appliquée, Institut de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium
| | - Delphine Lamoral-Theys
- Laboratoire de Toxicologie and Laboratoire de Chimie Analytique, Toxicologie et Chimie Physique Appliquée, Institut de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium
| | - Carla J. Hooten
- Department of Chemistry, New Mexico Institute of Mining and Technology, Socorro, New Mexico 87801, USA
| | - Pavel Y. Uglinskii
- Department of Organic Chemistry, Timiryazev Agriculture Academy, Moscow 127550, Russia
| | - Snezna Rogelj
- Department of Biology, New Mexico Institute of Mining and Technology, Socorro, New Mexico 87801, USA
| | - Robert Kiss
- Laboratoire de Toxicologie and Laboratoire de Chimie Analytique, Toxicologie et Chimie Physique Appliquée, Institut de Pharmacie, Université Libre de Bruxelles, Brussels, Belgium
| | - Wim F.A. Steelant
- Department of Chemistry, New Mexico Institute of Mining and Technology, Socorro, New Mexico 87801, USA
| | - Walter Berger
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| | - Jeremy J. Yang
- Division of Biocomputing, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Cristian G. Bologa
- Division of Biocomputing, Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Alexander Kornienko
- Department of Chemistry, New Mexico Institute of Mining and Technology, Socorro, New Mexico 87801, USA
| | - Igor V. Magedov
- Department of Chemistry, New Mexico Institute of Mining and Technology, Socorro, New Mexico 87801, USA
| |
Collapse
|
18
|
Cananzi S, Dallavalle S, Bargiotti A, Merlini L, Artali R, Beretta GL. Synthesis of new cytotoxic E-ring modified camptothecins. Tetrahedron Lett 2010. [DOI: 10.1016/j.tetlet.2010.09.130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
19
|
|
20
|
Affiliation(s)
- Yves Pommier
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4255, USA.
| |
Collapse
|
21
|
Basili S, Moro S. Novel camptothecin derivatives as topoisomerase I inhibitors. Expert Opin Ther Pat 2009; 19:555-74. [DOI: 10.1517/13543770902773437] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
22
|
Oostendorp RL, van de Steeg E, van der Kruijssen CMM, Beijnen JH, Kenworthy KE, Schinkel AH, Schellens JHM. Organic anion-transporting polypeptide 1B1 mediates transport of Gimatecan and BNP1350 and can be inhibited by several classic ATP-binding cassette (ABC) B1 and/or ABCG2 inhibitors. Drug Metab Dispos 2009; 37:917-23. [PMID: 19139163 DOI: 10.1124/dmd.108.024901] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Organic anion-transporting polypeptides (OATPs) are important uptake transporters that can have a profound impact on the systemic pharmacokinetics, tissue distribution, and elimination of several drugs. Previous in vivo studies of the pharmacokinetics of the lipophilic camptothecin (CPT) analog gimatecan suggested that the ATP-binding cassette (ABC) B1 (P-glycoprotein) and/or ABCG2 (breast cancer resistance protein) inhibitors elacridar and pantoprazole could inhibit transporters other than ABCB1 and ABCG2. In this study, we tested the possible role of OATP1B1 in this interaction by screening a number of CPT analogs for their transport affinity by human OATP1B1 in vitro. In addition, the impact of several widely used ABCB1 and/or ABCG2 modulators on this OATP1B1-mediated transport was assessed. We identified two novel CPT anticancer drugs, gimatecan and BNP1350, as OATP1B1 substrates, whereas irinotecan, topotecan, and lurtotecan were not transported by OATP1B1. It is interesting to note that transport of 17beta-estradiol 17beta-d-glucuronide (control), gimatecan, and BNP1350 by OATP1B1 could be completely inhibited by the classic ABCB1 and/or ABCG2 inhibitors elacridar, valspodar, pantoprazole, and, to a lesser extent, zosuquidar and verapamil. Therefore, the effect of these ABCB1 and ABCG2 modulators on the plasma pharmacokinetics of gimatecan and BNP1350 (and possibly also other OATP1B1 substrates) may be partly because of inhibition of OATP1B1 besides inhibition of ABCB1 and/or ABCG2. The findings of this study suggest that OATP1B1 polymorphisms or coadministration with one of the ABCB1/ABCG2 inhibitors could affect drug uptake, tissue distribution, and elimination of some CPT anticancer drugs, thereby modifying their efficacy and/or safety profile.
Collapse
Affiliation(s)
- Roos L Oostendorp
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
23
|
Beretta GL, Perego P, Zunino F. Targeting topoisomerase I: molecular mechanisms and cellular determinants of response to topoisomerase I inhibitors. Expert Opin Ther Targets 2008; 12:1243-56. [PMID: 18781823 DOI: 10.1517/14728222.12.10.1243] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Topoisomerase I is required for DNA relaxation during critical cellular functions. The identification of camptothecins as specific enzyme inhibitors and their clinical efficacy have stimulated extensive efforts to exploit topoisomerase I as a tumor target and explain the putative mechanisms of antitumor-specific action. OBJECTIVE This review provides an overview of the recent achievements in the development of topoisomerase I inhibitors and in the explanation of the biological pathways involved in tumor response. RESULTS/CONCLUSION In spite of the difficulty to identify novel topoisomerase I inhibitors with improved pharmacological properties, a growing body of evidence supports the possibility of optimizing the therapeutic profile of available agents. The explanation of defense mechanisms and the molecular determinants of tumor cell response is expected to provide a basis for the design of combination approaches for optimization of topoisomerase I inhibitors-based therapy.
Collapse
|
24
|
Takagi K, Dexheimer TS, Redon C, Sordet O, Agama K, Lavielle G, Pierré A, Bates SE, Pommier Y. Novel E-ring camptothecin keto analogues (S38809 and S39625) are stable, potent, and selective topoisomerase I inhibitors without being substrates of drug efflux transporters. Mol Cancer Ther 2008; 6:3229-38. [PMID: 18089716 DOI: 10.1158/1535-7163.mct-07-0441] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Camptothecin (CPT) analogues are powerful anticancer agents but are chemically unstable due to their alpha-hydroxylactone six-membered E-ring structure, which is essential for trapping topoisomerase I (Top1)-DNA cleavage complexes. To stabilize the E-ring, CPT keto analogues with a five-membered E-ring lacking the oxygen of the lactone ring (S38809 and S39625) have been synthesized. S39625 has been selected for advanced preclinical development based on its promising activity in tumor models. Here, we show that both keto analogues are active against purified Top1 and selective against Top1 in yeast and human cancer cells. The keto analogues show improved cytotoxicity toward colon, breast, and prostate cancer cells and leukemia cells compared with CPT. The drug-induced Top1-DNA cleavage complexes induced by the keto analogues show remarkable persistence both with purified Top1 and in cells following 1-h drug treatments. Moreover, we find that S39625 is not a substrate for either the ABCB1 (multidrug resistance-1/P-glycoprotein) or ABCG2 (mitoxantrone resistance/breast cancer resistance protein) drug efflux transporters, which sets S39625 apart from the clinically used CPT analogues topotecan or SN-38 (active metabolite of irinotecan). Finally, we show that nanomolar concentrations of S38809 or S39625 induce intense and persistent histone gamma-H2AX. The chemical stability of the keto analogues and the ability of S39625 to produce high levels of persistent Top1-DNA cleavage complex and its potent antiproliferative activity against human cancer cell lines make S39625 a promising new anticancer drug candidate. Histone gamma-H2AX could be used as a biomarker for the upcoming clinical trials of S39625.
Collapse
Affiliation(s)
- Kazutaka Takagi
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, 37 Convent Drive, Building 37, Room 5068, Bethesda, MD 20892-4255, USA
| | | | | | | | | | | | | | | | | |
Collapse
|