1
|
Cetinkaya A, Karadurmus L, Kaya SI, Ozcelikay G, Ozkan SA. Electrochemical Sensing of Anticancer Drug Using New Electrocatalytic Approach. Top Catal 2022. [DOI: 10.1007/s11244-021-01536-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
2
|
Duan YT, Sangani CB, Liu W, Soni KV, Yao Y. New Promises to Cure Cancer and Other Genetic Diseases/Disorders: Epi-drugs Through Epigenetics. Curr Top Med Chem 2019; 19:972-994. [DOI: 10.2174/1568026619666190603094439] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/05/2019] [Accepted: 05/27/2019] [Indexed: 12/13/2022]
Abstract
All the heritable alterations in gene expression and chromatin structure due to chemical modifications that do not involve changes in the primary gene nucleotide sequence are referred to as epigenetics. DNA methylation, histone modifications, and non-coding RNAs are distinct types of epigenetic inheritance. Epigenetic patterns have been linked to the developmental stages, environmental exposure, and diet. Therapeutic strategies are now being developed to target human diseases such as cancer with mutations in epigenetic regulatory genes using specific inhibitors. Within the past two decades, seven epigenetic drugs have received regulatory approval and many others show their candidature in clinical trials. The current article represents a review of epigenetic heritance, diseases connected with epigenetic alterations and regulatory approved epigenetic drugs as future medicines.
Collapse
Affiliation(s)
- Yong-Tao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou 450018, China
| | - Chetan B. Sangani
- Shri Maneklal M. Patel Institute of Sciences and Research, Kadi Sarva Vishwavidyalaya University, Gandhinagar, Gujarat, 362024, India
| | - Wei Liu
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou 450018, China
| | - Kunjal V. Soni
- Shri Maneklal M. Patel Institute of Sciences and Research, Kadi Sarva Vishwavidyalaya University, Gandhinagar, Gujarat, 362024, India
| | - Yongfang Yao
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
3
|
Pushkarev VM, SI V. P. Komisarenko Institute of Endocrinology and Metabolism, NAMS of Ukraine, Kyiv, Kovzun OI, Pushkarev VV, Guda BB, Tronko MD, SI V. P. Komisarenko Institute of Endocrinology and Metabolism, NAMS of Ukraine, Kyiv, SI V. P. Komisarenko Institute of Endocrinology and Metabolism, NAMS of Ukraine, Kyiv, SI V. P. Komisarenko Institute of Endocrinology and Metabolism, NAMS of Ukraine, Kyiv, SI V. P. Komisarenko Institute of Endocrinology and Metabolism, NAMS of Ukraine, Kyiv. Biochemical aspects of the combined use of taxanes, irradiation and other antineoplastic agents for the treatment of anaplastic thyroid carcinoma. UKRAINIAN BIOCHEMICAL JOURNAL 2018. [DOI: 10.15407/ubj90.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
4
|
Ma M, Zhao J, Cheng H, Deng M, Ding Z, Hou Y, Li F, Dou G, Li W. In vitro and in vivo pharmacokinetic and pharmacodynamic study of MBRI-001, a deuterium-substituted plinabulin derivative as a potent anti-cancer agent. Bioorg Med Chem 2018; 26:4687-4692. [PMID: 30119994 DOI: 10.1016/j.bmc.2018.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/26/2018] [Accepted: 08/04/2018] [Indexed: 02/06/2023]
Abstract
MBRI-001 was demonstrated preliminary better pharmacokinetics and antitumor effects than that of plinabulin in vivo. In this approach, we further carried out systematic pharmacokinetic and pharmacodynamic study of MBRI-001 in vitro and in vivo. MBRI-001 was tested stable in rat plasma and more stable in liver microsomes than plinabulin in vitro. In vivo, MBRI-001 could be distributed rapidly and widely in various tissues, especially the concentration of MBRI-001 in lung was remarkably higher than other tissues. Excretion study indicated that MBRI-001 might been decomposed and excreted as metabolites. Additionally, the combination treatment of MBRI-001 and gefitinib revealed better antitumor inhibition rate than monotherapy in vivo. Therefore, we suggest that MBRI-001 could be developed as a promising anti-cancer agent in near future.
Collapse
Affiliation(s)
- Mingxu Ma
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jianchun Zhao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Hejuan Cheng
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Mengyan Deng
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Zhongpeng Ding
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Yingwei Hou
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Feng Li
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Guifang Dou
- Institute of Transfusion Medicine, Academy of Military Medical Sciences, Beijing 100850, China
| | - Wenbao Li
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Innovation Center for Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China.
| |
Collapse
|
5
|
New insights into Vinca alkaloids resistance mechanism and circumvention in lung cancer. Biomed Pharmacother 2017; 96:659-666. [DOI: 10.1016/j.biopha.2017.10.041] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/08/2017] [Accepted: 10/09/2017] [Indexed: 12/22/2022] Open
|
6
|
The small molecule SI113 synergizes with mitotic spindle poisons in arresting the growth of human glioblastoma multiforme. Oncotarget 2017; 8:110743-110755. [PMID: 29340013 PMCID: PMC5762281 DOI: 10.18632/oncotarget.22500] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/29/2017] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the deadliest brain tumor. State-of-art GBM therapy often fails to ensure control of a disease characterized by high frequency of recurrences and progression. In search for novel therapeutic approaches, we assayed the effect of compounds from a cancer drug library on the ADF GBM cell line, establishing their elevated sensitivity to mitotic spindle poisons. Our previous work showed that the effectiveness of the spindle poison paclitaxel in inhibiting cancer cell growth was dependent on the expression of RANBP1, a regulatory target of the serine/threonine kinase SGK1. Recently, we developed the small molecule SI113 to inhibit SGK1 activity. Therefore, we explored the outcome of the association between SI113 and selected spindle poisons, finding that these drugs generated a synergistic cytotoxic effect in GBM cells, drastically reducing their viability and clonogenic capabilities in vitro, as well as inhibiting tumor growth in vivo. We also defined the molecular bases of such a synergistic effect. Because SI113 displays low systemic toxicity, yet strong activity in potentiating the effect of radiotherapy in GBM cells, we believe that this drug could be a strong candidate for clinical trials, with the aim to add it to the current GBM therapeutic approaches.
Collapse
|
7
|
Metformin and epothilone A treatment up regulate pro-apoptotic PARP-1, Casp-3 and H2AX genes and decrease of AKT kinase level to control cell death of human hepatocellular carcinoma and ovary adenocarcinoma cells. Toxicol In Vitro 2017; 47:48-62. [PMID: 29117515 DOI: 10.1016/j.tiv.2017.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/20/2017] [Accepted: 11/03/2017] [Indexed: 02/07/2023]
Abstract
High mortality rates in ovarian and liver cancer are largely a result of resistance to currently used chemotherapy. Here, we investigated genotoxic and pro-oxidant effects of metformin (MET) and epothilone A (A) in combination with respect to apoptosis in HepG2 and SKOV-3 cancer cells. Reactive oxygen species (ROS) was studied using 2',7'-dichlorodihydrofluoresein diacetate, and samples were analyzed for the presence and absence of the N-acetylcysteine (NAC). Expression of genes involved in programmed cell death, oxidative and alkylating DNA damage was measured. Probes were analyzed in the presence of Akt or nuclear factor-κB inhibitor. Compared to either drug alone, combination of epothilone A and metformin was more potent; decreased Akt level; and elevated percentage of apoptotic cells, induced cell cycle arrest at G1 phase and elevated the sub-G1 cell population by increasing the mRNA level of caspase-3, poly (ADP-ribose) polymerase-1 and H2AX. The anticancer effect of the drug combination was partially reversed by NAC supplementation, suggesting that ROS generation is required to induce apoptosis. The present study demonstrates that novel combination such as epothilone A and MET show promise in expanding ovarian and liver cancer therapy.
Collapse
|
8
|
Ziegler J, Bastian A, Lerner M, Bailey-Downs L, Saunders D, Smith N, Sutton J, Battiste JD, Ihnat MA, Gangjee A, Towner RA. AG488 as a therapy against gliomas. Oncotarget 2017; 8:71833-71844. [PMID: 29069750 PMCID: PMC5641093 DOI: 10.18632/oncotarget.18284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/05/2017] [Indexed: 11/25/2022] Open
Abstract
High-grade gliomas such as glioblastomas (GBM) present a deadly prognosis following diagnosis and very few effective treatment options. Here, we investigate if the small molecule AG488 can be an effective therapy against GBM with both anti-angiogenic as well as an anti-microtubule inhibiting modalities, using a human G55 glioma xenograft model in nude mice. From in vitro studies, we report that AG488 incubation reduced cell viability in G55 and HMEC-1 cells more so than TMZ treatment, and AG488 treatment also decreased cell viability in normal astrocytes, but not as much as for G55 cells (p<0.0001). In vivo investigations indicated that AG488 therapy helped reduce tumor volumes (p<0.0001), prolong survival (p<0.01), increase tumor perfusion (p<0.01), and decrease microvessel density (MVD) (p<0.05), compared to untreated mice or mice treated with non-specific IgG, in the G55 xenograft model. Additionally, AG488 did not induce apoptosis in normal mouse brain tissue. Animal survival and tumor volume changes for AG488 were comparable to TMZ or anti-VEGF therapies, however AG488 was found to be more effective in decreasing tumor-related vascularity (perfusion and MVD). AG488 is a potential novel therapy against high-grade gliomas.
Collapse
Affiliation(s)
- Jadith Ziegler
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.,Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anja Bastian
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Megan Lerner
- Department of Surgery Research Laboratory, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Lora Bailey-Downs
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jake Sutton
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - James D Battiste
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael A Ihnat
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Aleem Gangjee
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.,Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
9
|
Li YL, Sun J, Hu X, Pan YN, Yan W, Li QY, Wang F, Lin NM, Zhang C. Epothilone B induces apoptosis and enhances apoptotic effects of ABT-737 on human cancer cells via PI3K/AKT/mTOR pathway. J Cancer Res Clin Oncol 2016; 142:2281-9. [PMID: 27591861 DOI: 10.1007/s00432-016-2236-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 08/30/2016] [Indexed: 11/25/2022]
Abstract
PURPOSE Epothilone B and its derivatives are tested in multiple clinical trials. Epothilone B induces neurotoxic effect in clinical trials; however, low-dose epothilone B regimen can promote neuroprotection and neurogenesis. Thus, the study of new combination chemotherapy regimen incorporating low-dose epothilone B with other chemotherapeutic agents might help to develop epothilone B-based approaches to cancer treatment and avoid the neurotoxicity of epothilone B. METHODS Cell proliferation was assessed by SRB cell viability assay. Apoptosis was analyzed by propidium iodide (PI) staining. Mitochondrial membrane depolarization was evaluated using JC-1 staining. The expression of proteins was detected by western blotting. RESULTS In this study, we demonstrated that the combination of ABT-737 and low-dose epothilone B showed synergistic anti-proliferation effects on human cancer cells. In addition, epothilone B + ABT-737 synergy was through mitochondria-mediated apoptosis pathway. Furthermore, combination treatment markedly induced the activation of caspase-3 and the cleavage of PARP. The activation of PI3K/Akt/mTOR pathway is associated with resistance to epothilone B. Our data showed that epothilone B plus ABT-737 resulted in a blockade of the PI3K/AKT/mTOR signaling pathway. CONCLUSIONS These data indicate that ABT-737 may be a pertinent sensitizer to epothilone B, and the strategy of combining epothilone B with ABT-737 appears to be an attractive option for overcoming the resistance and neurotoxicity of epothilone B.
Collapse
Affiliation(s)
- Yang-Ling Li
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Nanjing Medical University, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Jiao Sun
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Xiu Hu
- School of Medicine, Zhejiang University City College, No. 51 Huzhou Street, Hangzhou, 310015, Zhejiang, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi-Ni Pan
- School of Medicine, Zhejiang University City College, No. 51 Huzhou Street, Hangzhou, 310015, Zhejiang, China
| | - Wei Yan
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Nanjing Medical University, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Qing-Yu Li
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Nanjing Medical University, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Fei Wang
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Nanjing Medical University, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China
| | - Neng-Ming Lin
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Nanjing Medical University, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China.
- Hangzhou Translational Medicine Research Center, Hangzhou First People's Hospital, Nanjing Medical University, No. 261 Huansha Road, Hangzhou, 310006, Zhejiang, China.
| | - Chong Zhang
- School of Medicine, Zhejiang University City College, No. 51 Huzhou Street, Hangzhou, 310015, Zhejiang, China.
| |
Collapse
|
10
|
Trendowski M. Using cytochalasins to improve current chemotherapeutic approaches. Anticancer Agents Med Chem 2015; 15:327-35. [PMID: 25322987 PMCID: PMC4485394 DOI: 10.2174/1871520614666141016164335] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 09/23/2014] [Accepted: 09/26/2014] [Indexed: 01/26/2023]
Abstract
Although the amount of progress cancer therapy has made in recent years is commendable, considerable limitations still remain. Most agents preferentially target rapidly proliferating cells, thereby destroying tumorigenic growths. Unfortunately, there are many labile cells in the patient that are also rapidly dividing, ultimately perpetuating significant side effects, including immunosuppression. Cytochalasins are microfilament-directed agents most commonly known for their use in basic research to understand cytoskeletal mechanisms. However, such agents also exhibit profound anticancer activity, as indicated by numerous in vitro and in vivo studies. Cytochalasins appear to preferentially damage malignant cells, as shown by their minimal effects on normal epithelial and immune cells. Further, cytochalasins influence the end stages of mitosis, suggesting that such agents could be combined with microtubule-directed agents to elicit a profound synergistic effect on malignant cells. Therefore, it is likely that cytochalasins could be used to supplement current chemotherapeutic measures to improve efficacy rates, as well as decrease the prevalence of drug resistance in the clinical setting.
Collapse
Affiliation(s)
- Matthew Trendowski
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA.
| |
Collapse
|
11
|
An F, Zhao WJ, Tang L, Qiu RG. Concentration-dependent differential effects of an epothilone analog on cell cycle and p53 signaling. Oncol Rep 2015; 34:1361-8. [PMID: 26177745 DOI: 10.3892/or.2015.4128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 06/22/2015] [Indexed: 11/06/2022] Open
Abstract
The tumor-suppressor protein p53 is considered to be one of the most important transport hubs of cell signal transduction, playing critical roles in the control of cell cycle arrest, apoptosis and many other processes as a nuclear transcription factor. p53 also acts in the cytoplasm to trigger apoptosis. Paclitaxel and other microtubule inhibitors can inhibit the growth of different types of cancer cells and induce apoptosis which is believed to be p53-independent. In the present study, we demonstrated that UTD1, a genetically engineered epothilone analog and a new microtubule inhibitor, activated p53 as a transcription factor at low concentrations demonstrated by its enhanced transcriptional activity and accumulation of p21, which led to cell cycle arrest. However, at high concentrations of UTD1, p53 was accumulated in the cytoplasm which contributed to induction of apoptosis. These observations indicate that the epothilone analog has differential effects on intracellular signaling and implies that p53 plays different roles in cells exposed to different concentrations of the anticancer agent.
Collapse
Affiliation(s)
- Fan An
- Research Center for Molecular Medicine, Dalian University of Technology, Dalian, Liaoning 116023, P.R. China
| | - Wei-Jie Zhao
- Research Center for Molecular Medicine, Dalian University of Technology, Dalian, Liaoning 116023, P.R. China
| | - Li Tang
- Research Center for Molecular Medicine, Dalian University of Technology, Dalian, Liaoning 116023, P.R. China
| | - Rong-Guo Qiu
- Research Center for Molecular Medicine, Dalian University of Technology, Dalian, Liaoning 116023, P.R. China
| |
Collapse
|
12
|
Hassan MK, Watari H, Mitamura T, Mohamed Z, EL-khamisy SF, Ohba Y, Sakuragi N. P18/Stathmin1 is regulated by miR-31 in ovarian cancer in response to taxane. Oncoscience 2015; 2:294-308. [PMID: 25897432 PMCID: PMC4394135 DOI: 10.18632/oncoscience.143] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 03/16/2015] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRNAs) have been reported to regulate the development of chemoresistance in many tumors. Stathmin 1 (STMN1) is a microtubule-depolymerizing molecule, involved in chemo-response; however, the mechanism of its regulation is unknown. Herein, the immunohistochemical study indicated significant upregulation of the STMN1 in the ovarian cancer tissues defined as resistant tumors compared with those defined as responsive tumors. STMN1 level elevated in the chemoresistant ovarian cancer cells, KF-TX, compared with the parental, KF, ones. Targeting STMN1 by siRNA restored taxane-sensitivity of KF-TX cells. Screening miRNA profiles from KF/KF-TX cellular set followed by bioinformatics-based prediction, revealed that miR-31 could be a possible regulator of STMN1. Down-modulation of miR-31 was verified by quantitative RT-PCR in the cellular set used. Overexpression of miR-31 in KF-TX cells (KF-TX-miR-31) significantly restored chemo-response and reduced STMN1 expression as well. STMN1 reduction-associated cellular characteristics such as enhanced microtubule polymerization and stability, as indicated by acetylated tubulin quantification, confocal visualization, and G2 phase delay, were observed in KF-TX-miR-31 cells, indicating the functional reduction of STMN1. miR-31 suppressed the luciferase activity in reporter construct containing the STMN1 3'-untranslated region (3'-UTR), confirming that miR-31 directly targets STMN1. miR-31 has therapeutic potency when introduced into ovarian cancer, in combination with taxane.
Collapse
Affiliation(s)
- Mohamed Kamel Hassan
- Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo, JAPAN
- Bitechnology Program, Zoology Department, Faculty of Science, Port Said University, Port Said, EGYPT
- Center of Genomics, Hemly Institute for Medical Sciences, Zewail City for Science and Technology, Giza, EGYPT
| | - Hidemichi Watari
- Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo, JAPAN
| | - Takashi Mitamura
- Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo, JAPAN
| | - Zainab Mohamed
- Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo, JAPAN
| | - Sherif F. EL-khamisy
- Center of Genomics, Hemly Institute for Medical Sciences, Zewail City for Science and Technology, Giza, EGYPT
- Krebs Institute, Department of Molecular Biology and Biotechnology, University of Sheffield, UK
| | - Yusuke Ohba
- Department of Cell Physiology, Hokkaido University Graduate School of Medicine, Sapporo, JAPAN
| | - Noriaki Sakuragi
- Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo, JAPAN
| |
Collapse
|
13
|
Eichhorn JM, Kothari A, Chambers TC. Cyclin B1 overexpression induces cell death independent of mitotic arrest. PLoS One 2014; 9:e113283. [PMID: 25415322 PMCID: PMC4240608 DOI: 10.1371/journal.pone.0113283] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/21/2014] [Indexed: 11/23/2022] Open
Abstract
Microtubule inhibitors are widely used in cancer chemotherapy. These drugs characteristically induce mitotic arrest and cell death but the mechanisms linking the two are not firmly established. One of the problems is that cancer cells vary widely in their sensitivity to these agents, and thus comparison of data from different systems is difficult. To alleviate this problem we sought to molecularly induce mitotic death and study its mechanisms, by expressing non-degradable cyclin B (R42A) in HeLa cells. However, this approach failed to induce significant mitotic arrest, Cdk1 activation, or phosphorylation of anti-apoptotic Bcl-2 proteins, all characteristics of cells treated with microtubule inhibitors. Furthermore, cyclin B1-R42A induced rapid cell death, and when expressed in synchronized cells, cell death occurred in G1 phase. Decreasing the plasmid concentration reduced transfection efficiency but restored mitotic arrest and eliminated non-specific death. These results show that inappropriate overexpression of cyclin B1 causes non-specific cell death and suggest caution in its use for the study of mitotic events.
Collapse
Affiliation(s)
- Joshua M. Eichhorn
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arizona, United States of America
| | - Anisha Kothari
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arizona, United States of America
| | - Timothy C. Chambers
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arizona, United States of America
| |
Collapse
|
14
|
Trendowski M. Exploiting the cytoskeletal filaments of neoplastic cells to potentiate a novel therapeutic approach. Biochim Biophys Acta Rev Cancer 2014; 1846:599-616. [PMID: 25286320 DOI: 10.1016/j.bbcan.2014.09.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 09/19/2014] [Accepted: 09/21/2014] [Indexed: 02/06/2023]
Abstract
Although cytoskeletal-directed agents have been a mainstay in chemotherapeutic protocols due to their ability to readily interfere with the rapid mitotic progression of neoplastic cells, they are all microtubule-based drugs, and there has yet to be any microfilament- or intermediate filament-directed agents approved for clinical use. There are many inherent differences between the cytoskeletal networks of malignant and normal cells, providing an ideal target to attain preferential damage. Further, numerous microfilament-directed agents, and an intermediate filament-directed agent of particular interest (withaferin A) have demonstrated in vitro and in vivo efficacy, suggesting that cytoskeletal filaments may be exploited to supplement chemotherapeutic approaches currently used in the clinical setting. Therefore, this review is intended to expose academics and clinicians to the tremendous variety of cytoskeletal filament-directed agents that are currently available for further chemotherapeutic evaluation. The mechanisms by which microfilament directed- and intermediate filament-directed agents damage malignant cells are discussed in detail in order to establish how the drugs can be used in combination with each other, or with currently approved chemotherapeutic agents to generate a substantial synergistic attack, potentially establishing a new paradigm of chemotherapeutic agents.
Collapse
Affiliation(s)
- Matthew Trendowski
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA.
| |
Collapse
|
15
|
Zhang X, Raghavan S, Ihnat M, Thorpe JE, Disch BC, Bastian A, Bailey-Downs LC, Dybdal-Hargreaves NF, Rohena CC, Hamel E, Mooberry SL, Gangjee A. The design and discovery of water soluble 4-substituted-2,6-dimethylfuro[2,3-d]pyrimidines as multitargeted receptor tyrosine kinase inhibitors and microtubule targeting antitumor agents. Bioorg Med Chem 2014; 22:3753-72. [PMID: 24890652 PMCID: PMC4089508 DOI: 10.1016/j.bmc.2014.04.049] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/16/2014] [Accepted: 04/25/2014] [Indexed: 12/13/2022]
Abstract
The design, synthesis and biological evaluations of fourteen 4-substituted 2,6-dimethylfuro[2,3-d]pyrimidines are reported. Four compounds (11-13, 15) inhibit vascular endothelial growth factor receptor-2 (VEGFR-2), platelet-derived growth factor receptor β (PDGFR-β), and target tubulin leading to cytotoxicity. Compound 11 has nanomolar potency, comparable to sunitinib and semaxinib, against tumor cell lines overexpressing VEGFR-2 and PDGFR-β. Further, 11 binds at the colchicine site on tubulin, depolymerizes cellular microtubules and inhibits purified tubulin assembly and overcomes both βIII-tubulin and P-glycoprotein-mediated drug resistance, and initiates mitotic arrest leading to apoptosis. In vivo, its HCl salt, 21, reduced tumor size and vascularity in xenograft and allograft murine models and was superior to docetaxel and sunitinib, without overt toxicity. Thus 21 affords potential combination chemotherapy in a single agent.
Collapse
Affiliation(s)
- Xin Zhang
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States
| | - Sudhir Raghavan
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States
| | - Michael Ihnat
- College of Pharmacy, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Jessica E Thorpe
- College of Pharmacy, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Bryan C Disch
- College of Pharmacy, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Anja Bastian
- College of Pharmacy, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Lora C Bailey-Downs
- College of Pharmacy, University of Oklahoma Health Science Center, 1110 North Stonewall, Oklahoma City, OK 73117, United States
| | - Nicholas F Dybdal-Hargreaves
- Department of Pharmacology, Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Cristina C Rohena
- Department of Pharmacology, Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Institutes of Health, 1050 Boyles Street, Frederick, MD 21702, United States
| | - Susan L Mooberry
- Department of Pharmacology, Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Aleem Gangjee
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, Pittsburgh, PA 15282, United States.
| |
Collapse
|
16
|
Wu YK, Huang CY, Yang MC, Lan CC, Lee CH, Chan EC, Chen KT. Nuclear survivin expression: a prognostic factor for the response to taxane-platinum chemotherapy in patients with advanced non-small cell lung cancer. Med Oncol 2014; 31:79. [PMID: 24961465 DOI: 10.1007/s12032-014-0079-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 06/13/2014] [Indexed: 01/17/2023]
Abstract
Survivin, a structurally unique protein expressed in most common human neoplasms, is thought to support cell cycle progression and suppress apoptosis. Survivin expression is highly correlated with advanced non-small cell lung cancer (NSCLC) and poor prognosis. In this retrospective study of banked pathology tissue of patients with advanced NSCLC, we tested for correlations of N-survivin expression in tumor tissues and responsiveness to treatment with platinum-based regimens containing paclitaxel or docetaxel. The 48 patients with NSCLC included 32 (66.7 %) males and 16 (33.3 %) females. Mean age at diagnosis was 59.4 years (range 36-83 years), and median follow-up time was 20.4 months (range 3.4-59.0 months). Patients with high tumor N-survivin expression had significantly better responses to taxane-platinum chemotherapy than those with low tumor N-survivin expression (P < 0.001). Adjusted multivariate modeling found high tumor N-survivin expression to be an independent prognostic factor for a clinical response to chemotherapy (high vs. low, OR 6.14, 95 % CI 1.62-23.29; P = 0.008). Median overall survival differed significantly between those with high tumor N-survivin expression who did/did not respond to chemotherapy and between those with low tumor N-survivin expression who did/did not respond to chemotherapy (P < 0.05). Tumor N-survivin expression shows promise as a predictive biomarker in the chemotherapy setting as a surrogate marker of high proliferation status.
Collapse
Affiliation(s)
- Yao-Kuang Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
17
|
Frye DK. Advances in breast cancer treatment: the emerging role of ixabepilone. Expert Rev Anticancer Ther 2014; 10:23-32. [DOI: 10.1586/era.09.158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
18
|
Song H, Zhou S, Wang R, Li S. Kinesin spindle protein (KSP) inhibitors in combination with chemotherapeutic agents for cancer therapy. ChemMedChem 2013; 8:1736-49. [PMID: 23964020 DOI: 10.1002/cmdc.201300228] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/20/2013] [Indexed: 12/20/2022]
Abstract
A diverse group of proteins, the activities of which are precisely orchestrated during mitosis, have emerged as targets for cancer therapeutics; these include the Aurora kinases (AKs), Polo-like kinases (PLKs), and the kinesin spindle protein (KSP). KSP is essential for the proper separation of spindle poles during mitosis. Agents that target KSP selectively act on cells undergoing cell division, which means that KSP inhibitors are mitosis-specific drugs, and have demonstrated remarkable activities in vitro. However, a significant obstacle to the success of KSP inhibitors is that these compounds, with tremendous efficacy in vitro, have demonstrated little or even no antitumor activity in vivo. Accumulated data suggest that a combination of KSP inhibitors with various cytostatic drugs will result in a more powerful tumor-killing effect than monotherapy. Combination therapies might predominate and represent the next frontier in the discovery research of KSP inhibitors as potential anticancer drugs. Few published studies have reviewed combination therapy using KSP inhibitors. Herein we provide a comprehensive review of the literature on KSP inhibitor monotherapy and therapeutic combinations. The current state and problems are also discussed.
Collapse
Affiliation(s)
- Hualong Song
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai (PR China)
| | | | | | | |
Collapse
|
19
|
Apoptosis induced by paclitaxel via Bcl-2, Bax and caspases 3 and 9 activation in NB4 human leukaemia cells is not modulated by ERK inhibition. ACTA ACUST UNITED AC 2013; 65:1101-8. [PMID: 23735541 DOI: 10.1016/j.etp.2013.04.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 03/25/2013] [Accepted: 04/25/2013] [Indexed: 11/22/2022]
Abstract
We have studied the role of pivotal bio-molecules involved in signalling of cytotoxic effects induced by paclitaxel (Ptx) on acute promyelocytic human leukaemia NB4 cells. A time-dependent increase in cell death and DNA cleavage was observed after 30μM Ptx treatment. Cell death induction by Ptx proceeds mainly as programmed cell death as shown by annexin V-FITC, reaching up to 30% of apoptotic cells after 24h. Significant reductions of p53, changes in Bax and Bcl-2 and activation of caspases 3 and 9 were observed as the treatment was applied for long times. Ptx treatments produced NFkB depletion with expression levels abolished at 19h what could be involved in reduction of survival signals. Phosphorylation of intracellular kinases showed that pERK1/2 decreased significantly at 19h of Ptx treatment. When these cells were preincubated for 90min with 20μM PD98059, 2'-amino-3'-methoxyflavone, an inhibitor of ERK phosphorylation, a slight reduction of cell viability was observed in comparison to that produced by Ptx alone. Pretreatment with PD98059 neither activated caspases nor significantly increased the apoptotic effect of Ptx. Taken together, our data reveal that the inhibition of ERK phosphorylation does not seem to be an essential pathway for bursting an increased induction of apoptosis by Ptx. Decrease of p53 and Bcl-2, fragmentation of DNA, increase of Bax and, finally, activation of caspases 3 and 9 in NB4 leukaemia cells make the apoptotic process induced by Ptx irreversible. Application of Ptx in leukaemia cells shows therefore a promising potential with particular effects on different leukaemia cell types.
Collapse
|
20
|
DaSilva JO, Amorino GP, Casarez EV, Pemberton B, Parsons SJ. Neuroendocrine-derived peptides promote prostate cancer cell survival through activation of IGF-1R signaling. Prostate 2013; 73. [PMID: 23192379 PMCID: PMC4085781 DOI: 10.1002/pros.22624] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Neuroendocrine (NE) cells promote the progression of prostate cancer to a castration-resistant state through the production of paracrine growth factors. We have demonstrated this principle using in vitro and in vivo proliferative endpoints; however, the contributions of NE-derived pro-survival factors and anti-apoptosis to this phenomenon have not been thoroughly investigated. METHODS Here, we utilized conditioned-medium (CM) from LNCaP cells, engineered to undergo NE differentiation, and examined its effects on PC3 and LNCaP cell survival. RESULTS Statistically significant changes in clonogenic survival, Annexin V staining, PARP cleavage and trypan blue positivity of approximately twofold were observed in the presence of NE-derived CM relative to control-CM for both LNCaP and PC3 cells. These changes were partially abrogated by antagonists of the neuropeptides neurotensin, bombesin, and PTHrP. Selective inhibitors of IGF-1R, EGFR or Src caused significant and nearly complete blockade of prostate cancer cell survival due to NE secretions. Similar increases in cell survival were observed for LNCaP or PC3 cells treated with NE-derived medium in the presence of docetaxel. Increased phosphorylation of IGF-1R, following treatment with NE-derived medium, was accompanied by decreased protein tyrosine phosphatase, receptor type F (PTPRF) mRNA, and protein levels. Overexpression of PTPRF decreased cell survival, the amplitude and duration of IGF-1R phosphorylation, and enhanced PARP cleavage in the presence of NE-derived medium. CONCLUSIONS These data support the hypothesis that NE-derived factors act upon prostate cancer cells to stimulate pro-survival signaling and describe a novel mechanism of cross-talk between NE-derived factors and IGF-1R, mediated in part by PTPRF.
Collapse
MESH Headings
- Blotting, Western
- Cell Line, Tumor
- Cell Survival/physiology
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/metabolism
- Gene Expression Regulation, Enzymologic
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Neoplasms, Hormone-Dependent/enzymology
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/metabolism
- Neurosecretory Systems/metabolism
- Parathyroid Hormone-Related Protein/antagonists & inhibitors
- Parathyroid Hormone-Related Protein/metabolism
- Prostatic Neoplasms/enzymology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- RNA, Messenger/chemistry
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/metabolism
- Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics
- Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- John O. DaSilva
- Departments of Microbiology, Cancer Center, University of Virginia Health Sciences Center, Charlottesville, VA 22908
| | - George P. Amorino
- Radiation Oncology and Cancer Center, University of Virginia Health Sciences Center, Charlottesville, VA 22908
| | - Eli V. Casarez
- Departments of Microbiology, Cancer Center, University of Virginia Health Sciences Center, Charlottesville, VA 22908
| | - Bradley Pemberton
- Radiation Oncology and Cancer Center, University of Virginia Health Sciences Center, Charlottesville, VA 22908
| | - Sarah J. Parsons
- Departments of Microbiology, Cancer Center, University of Virginia Health Sciences Center, Charlottesville, VA 22908
| |
Collapse
|
21
|
Inhibition of SIRT2 potentiates the anti-motility activity of taxanes: implications for antineoplastic combination therapies. Neoplasia 2013; 14:846-54. [PMID: 23019416 DOI: 10.1593/neo.12728] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 08/06/2012] [Accepted: 08/06/2012] [Indexed: 12/11/2022] Open
Abstract
Taxanes are potent inhibitors of cell motility, a property implicated in their antiangiogenic and antimetastatic activity and unrelated to their antiproliferative effect. The molecular mechanism of this anti-motility activity is poorly understood. In this study, we found that paclitaxel induced tubulin acetylation in endothelial and tumor cells, at concentrations that affected cell motility but not proliferation (10(-8) to 10(-9) M, for 4 hours). Induction of tubulin acetylation correlated with inhibition of motility but not proliferation based on a comparison of highly and poorly cytotoxic taxanes (paclitaxel and IDN5390) and tumor cell lines sensitive and resistant to paclitaxel (1A9 and 1A9 PTX22). Consistent with the hypothesis that tubulin deacetylase activity might affect cell response to the anti-motility activity of taxanes, we found that overexpression of the tubulin deacetylase SIRT2 increased cell motility and reduced cell response to the anti-motility activity of paclitaxel. Conversely, the SIRT2 inhibitor splitomicin reduced cell motility and potentiated the anti-motility activity of paclitaxel. The inhibitory effect was further potentiated by the addition of the HDAC6 inhibitor trichostatin A. Paclitaxel and splitomicin promoted translocation into the nucleus--and hence activation--of FOXO3a, a negative regulator of cell motility. This study indicates a role for SIRT2 in the regulation of cell motility and suggests that therapies combining sirtuin inhibitors and taxanes could be used to treat cell motility-based pathologic processes such as tumor angiogenesis, invasion, and metastasis.
Collapse
|
22
|
Docetaxel and 5-fluorouracil induce human p53 tumor suppressor gene transcription via a short sequence at core promoter element. Toxicol In Vitro 2012; 26:678-85. [DOI: 10.1016/j.tiv.2012.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2011] [Revised: 02/24/2012] [Accepted: 03/07/2012] [Indexed: 11/19/2022]
|
23
|
Morikawa Y, Koike H, Sekine Y, Matsui H, Shibata Y, Ito K, Suzuki K. Rapamycin enhances docetaxel-induced cytotoxicity in a androgen-independent prostate cancer xenograft model by survivin downregulation. Biochem Biophys Res Commun 2012; 419:584-9. [DOI: 10.1016/j.bbrc.2012.02.089] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 02/15/2012] [Indexed: 10/28/2022]
|
24
|
Pushkarev VV, Kovzun OI, Pushkarev VM, Tronko MD. The effect of the combined action of roscovitine and Paclitaxel on the apoptotic and cell cycle regulatory mechanisms in colon and anaplastic thyroid cancer cells. ISRN BIOCHEMISTRY 2012; 2012:826305. [PMID: 25969761 PMCID: PMC4392990 DOI: 10.5402/2012/826305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 08/07/2012] [Indexed: 11/24/2022]
Abstract
Aim. To study the significance of cyclin-dependent kinases (Cdks) in paclitaxel-dependent apoptosis in colon and undifferentiated thyroid cancer cells. Materials and Methods. Experiments were performed on undifferentiated thyroid carcinoma (KTC-2) and colon carcinoma (ARO) cell lines. Cells were treated with paclitaxel (Ptx) and inhibitor of Cdk, roscovitine. Cell survival test and Western blotting were used for characterization of the effects of paclitaxel and roscovitine on cancer cells. Results. It was shown that not c-Jun N-terminal kinase, but cyclin-dependent kinases are responsible for antiapoptotic Bcl-2 phosphorylation. Cdk inhibition enhanced the cytotoxic effects of Ptx at low drug concentrations. There was antagonism between Ptx and roscovitine at higher (25 nM) paclitaxel concentrations. Conclusion. Using of paclitaxel at low (2.5 to 5 nM) concentrations and roscovitine is a promising combination for further preclinical trials for the development of new therapeutic approaches to the treatment of colon and anaplastic thyroid cancer.
Collapse
Affiliation(s)
- V V Pushkarev
- Department of Fundamental Problems of Endocrinology, State Institution "V.P. Komisarenko Institute of Endocrinology and Metabolism", AMS of Ukraine, Kyiv 04114, Ukraine
| | - O I Kovzun
- Department of Fundamental Problems of Endocrinology, State Institution "V.P. Komisarenko Institute of Endocrinology and Metabolism", AMS of Ukraine, Kyiv 04114, Ukraine
| | - V M Pushkarev
- Department of Fundamental Problems of Endocrinology, State Institution "V.P. Komisarenko Institute of Endocrinology and Metabolism", AMS of Ukraine, Kyiv 04114, Ukraine
| | - M D Tronko
- Department of Fundamental Problems of Endocrinology, State Institution "V.P. Komisarenko Institute of Endocrinology and Metabolism", AMS of Ukraine, Kyiv 04114, Ukraine
| |
Collapse
|
25
|
Eschenbrenner J, Winsel S, Hammer S, Sommer A, Mittelstaedt K, Drosch M, Klar U, Sachse C, Hannus M, Seidel M, Weiss B, Merz C, Siemeister G, Hoffmann J. Evaluation of activity and combination strategies with the microtubule-targeting drug sagopilone in breast cancer cell lines. Front Oncol 2011; 1:44. [PMID: 22649765 PMCID: PMC3355879 DOI: 10.3389/fonc.2011.00044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 10/31/2011] [Indexed: 12/14/2022] Open
Abstract
Sagopilone, a fully synthetic epothilone, is a microtubule-stabilizing agent optimized for high in vitro and in vivo activity against a broad range of tumor models, including those resistant to paclitaxel and other systemic treatments. Sagopilone development is accompanied by translational research studies to evaluate the molecular mode of action, to recognize mechanisms leading to resistance, to identify predictive response biomarkers, and to establish a rationale for combination with different therapies. Here, we profiled sagopilone activity in breast cancer cell lines. To analyze the mechanisms of mitotic arrest and apoptosis and to identify additional targets and biomarkers, an siRNA-based RNAi drug modifier screen interrogating 300 genes was performed in four cancer cell lines. Defects of the spindle assembly checkpoint (SAC) were identified to cause resistance against sagopilone-induced mitotic arrest and apoptosis. Potential biomarkers for resistance could therefore be functional defects like polymorphisms or mutations in the SAC, particularly in the central SAC kinase BUB1B. Moreover, chromosomal heterogeneity and polyploidy are also potential biomarkers of sagopilone resistance since they imply an increased tolerance for aberrant mitosis. RNAi screening further demonstrated that the sagopilone-induced mitotic arrest can be enhanced by concomitant inhibition of mitotic kinesins, thus suggesting a potential combination therapy of sagopilone with a KIF2C (MCAK) kinesin inhibitor. However, the combination of sagopilone and inhibition of the prophase kinesin KIF11 (EG5) is antagonistic, indicating that the kinesin inhibitor has to be highly specific to bring about the required therapeutic benefit.
Collapse
Affiliation(s)
- Julia Eschenbrenner
- Global Drug Discovery, Therapeutic Research Group Oncology, Bayer Healthcare Pharmaceuticals Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Felisiak-Golabek A, Rembiszewska A, Rzepecka IK, Szafron L, Madry R, Murawska M, Napiorkowski T, Sobiczewski P, Osuch B, Kupryjanczyk J. Nuclear survivin expression is a positive prognostic factor in taxane-platinum-treated ovarian cancer patients. J Ovarian Res 2011; 4:20. [PMID: 22075440 PMCID: PMC3223127 DOI: 10.1186/1757-2215-4-20] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 11/10/2011] [Indexed: 11/16/2022] Open
Abstract
Background Survivin is an inhibitor of apoptosis and a regulator of mitotic progression. TP53 protein is a negative transcriptional regulator of survivin. The aim of our study was to evaluate the clinical significance of survivin expression in advanced stages ovarian cancer with respect to the TP53 status. Methods Survivin and TP53 expression was evaluated immunohistochemically in 435 archival samples of ovarian carcinomas (244 patients were treated with platinum/cyclophosphamide-PC/PAC; 191-with taxane-platinum (TP) agents). Univariate and multivariate statistical analyses were performed in patients groups divided according to the administered chemotherapeutic regimen, and in subgroups with and without TP53 accumulation (TP53+ and TP53-, respectively). Results Nuclear and cytoplasmic survivin expression was observed in 92% and 74% of the carcinomas, respectively. In patients treated with TP, high nuclear survivin expression decreased the risk of disease recurrence and death, and increased the probability of high platinum sensitivity (p < 0.01), but only in the TP53(+) group, and not in the TP53(-) group. Conclusions It appears that TP53 status determines the clinical importance of nuclear survivin expression in taxane-platinum treated ovarian cancer patients.
Collapse
Affiliation(s)
- Anna Felisiak-Golabek
- Department of Molecular Pathology, The Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Froidevaux-Klipfel L, Poirier F, Boursier C, Crépin R, Poüs C, Baudin B, Baillet A. Modulation of septin and molecular motor recruitment in the microtubule environment of the Taxol-resistant human breast cancer cell line MDA-MB-231. Proteomics 2011; 11:3877-86. [PMID: 21761557 DOI: 10.1002/pmic.201000789] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 06/08/2011] [Accepted: 07/05/2011] [Indexed: 01/09/2023]
Abstract
Cell resistance to low doses of paclitaxel (Taxol) involves a modulation of microtubule (MT) dynamics. We applied a proteomic approach based on 2-DE coupled with MS to identify changes in the MT environment of Taxol-resistant breast cancer cells. Having established a proteomic pattern of the microtubular proteins extracted from MDA-MB-231 cells, we verified by Western blotting that in resistant cells, α- and β-tubulins (more specifically the βIII and βIV isotypes) increased. Interestingly, four septins (SEPT2, 8, 9 and 11), which are GTPases involved in cytokinesis and in MT/actin cytoskeleton organization, were overexpressed and enriched in the MT environment of Taxol-resistant cells compared to their sensitive counterpart. Changes in the MT proteome of resistant cells also comprised increased kinesin-1 heavy chain expression and recruitment on MTs while dynein light chain-1 was downregulated. Modulation of motor protein recruitment around MTs might reflect their important role in controlling MT dynamics via the organization of signaling pathways. The identification of proteins previously unknown to be linked to taxane-resistance could also be valuable to identify new biological markers of resistance.
Collapse
|
28
|
|
29
|
Gertsch J, Viveros-Paredes JM, Taylor P. Plant immunostimulants--scientific paradigm or myth? JOURNAL OF ETHNOPHARMACOLOGY 2011; 136:385-391. [PMID: 20620205 DOI: 10.1016/j.jep.2010.06.044] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 05/19/2010] [Accepted: 06/23/2010] [Indexed: 05/29/2023]
Abstract
In traditional medicine, numerous plant preparations are used to treat inflammation both topically and systemically. Several anti-inflammatory plant extracts and a few natural product-based monosubstances have even found their way into the clinic. Unfortunately, a number of plant secondary metabolites have been shown to trigger detrimental pro-allergic immune reactions and are therefore considered to be toxic. In the phytotherapy research literature, numerous plants are also claimed to exert immunostimulatory effects. However, while the concepts of plant-derived anti-inflammatory agents and allergens are well established, the widespread notion of immunostimulatory plant natural products and their potential therapeutic use is rather obscure, often with the idea that the product is some sort of "tonic" for the immune system without actually specifying the mechanisms. In this commentary it is argued that the paradigm of oral plant immunostimulants lacks clinical evidence and may therefore be a myth, which has originated primarily from in vitro studies with plant extracts. The fact that no conclusive data on orally administered immunostimulants can be found in the scientific literature inevitably prompts us to challenge this paradigm.
Collapse
Affiliation(s)
- Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, Bern, Switzerland.
| | | | | |
Collapse
|
30
|
Rescue of neurons from undergoing hallmark tau-induced Alzheimer's disease cell pathologies by the antimitotic drug paclitaxel. Neurobiol Dis 2011; 43:163-75. [DOI: 10.1016/j.nbd.2011.03.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 02/14/2011] [Accepted: 03/06/2011] [Indexed: 11/18/2022] Open
|
31
|
Samadi N, Bekele RT, Goping IS, Schang LM, Brindley DN. Lysophosphatidate induces chemo-resistance by releasing breast cancer cells from taxol-induced mitotic arrest. PLoS One 2011; 6:e20608. [PMID: 21647386 PMCID: PMC3103588 DOI: 10.1371/journal.pone.0020608] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 05/05/2011] [Indexed: 11/18/2022] Open
Abstract
Background Taxol is a microtubule stabilizing agent that arrests cells in mitosis leading to cell death. Taxol is widely used to treat breast cancer, but resistance occurs in 25–69% of patients and it is vital to understand how Taxol resistance develops to improve chemotherapy. The effects of chemotherapeutic agents are overcome by survival signals that cancer cells receive. We focused our studies on autotaxin, which is a secreted protein that increases tumor growth, aggressiveness, angiogenesis and metastasis. We discovered that autotaxin strongly antagonizes the Taxol-induced killing of breast cancer and melanoma cells by converting the abundant extra-cellular lipid, lysophosphatidylcholine, into lysophosphatidate. This lipid stimulates specific G-protein coupled receptors that activate survival signals. Methodology/Principal Findings In this study we determined the basis of these antagonistic actions of lysophosphatidate towards Taxol-induced G2/M arrest and cell death using cultured breast cancer cells. Lysophosphatidate does not antagonize Taxol action in MCF-7 cells by increasing Taxol metabolism or its expulsion through multi-drug resistance transporters. Lysophosphatidate does not lower the percentage of cells accumulating in G2/M by decreasing exit from S-phase or selective stimulation of cell death in G2/M. Instead, LPA had an unexpected and remarkable action in enabling MCF-7 and MDA-MB-468 cells, which had been arrested in G2/M by Taxol, to normalize spindle structure and divide, thus avoiding cell death. This action involves displacement of Taxol from the tubulin polymer fraction, which based on inhibitor studies, depends on activation of LPA receptors and phosphatidylinositol 3-kinase. Conclusions/Significance This work demonstrates a previously unknown consequence of lysophosphatidate action that explains why autotaxin and lysophosphatidate protect against Taxol-induced cell death and promote resistance to the action of this important therapeutic agent.
Collapse
Affiliation(s)
- Nasser Samadi
- Department of Biochemistry (Signal Transduction Research Group), School of Molecular and Systems Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Raie T. Bekele
- Department of Biochemistry (Signal Transduction Research Group), School of Molecular and Systems Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Ing Swie Goping
- Department of Biochemistry (Signal Transduction Research Group), School of Molecular and Systems Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Luis M. Schang
- Department of Biochemistry (Signal Transduction Research Group), School of Molecular and Systems Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - David N. Brindley
- Department of Biochemistry (Signal Transduction Research Group), School of Molecular and Systems Medicine, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
32
|
Wang F, Wang LX, Li SL, Li K, He W, Liu HT, Fan QX. Downregulation of stathmin is involved in malignant phenotype reversion and cell apoptosis in esophageal squamous cell carcinoma. J Surg Oncol 2011; 103:704-15. [PMID: 21360534 DOI: 10.1002/jso.21870] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2010] [Accepted: 01/03/2011] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND OBJECTIVES Stathmin plays a critical role in the regulation of mitosis and mediates the development of malignant tumors. Here, we investigated the potential role of stathmin in cell cycle and apoptosis in esophageal squamous cell carcinoma (ESCC). METHODS A stathmin short hairpin RNA (shRNA) plasmid was employed to downregulate stathmin expression in the ESCC cell line EC9706 cells. Cell proliferation was measured by cell counting, MTT, and colony formation assay. Cell migration was measured by Boyden chamber. Western blot was used to analyze the expressions of stathmin, survivin, and apoptosis-related proteins in transfected cells. Cell cycle and apoptosis were determined by flow cytometry and DNA ladder. Oncogenicity assay in nude mice was utilized to analyze phenotypic changes of transfected cells in vivo. RESULTS After transfection with stathmin shRNA plasmid, stathmin expression markedly decreased in EC9706 cells. Stathmin downregulation significantly inhibited cell proliferation, cell migration in vitro, and tumorigenicity in vivo, meanwhile arrested cell cycle in the G2/M phase and induced cell apoptosis. Further, stathmin downregulation resulted in downregulation of Bcl-2 and survivin proteins, activation of Caspase-3. CONCLUSIONS These findings demonstrate that stathmin may play an essential role in carcinogenesis of ESCC, which will lay a foundation for target therapy of ESCC.
Collapse
Affiliation(s)
- Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
IMPORTANCE OF THE FIELD Since the introduction of taxane-based chemotherapy for advanced solid tumors in the 1990s, the median overall survival of patients with metastatic breast cancer increased from 2 years to almost 5 years. Similarly, the 5-year overall survival for ovarian cancer has increased from 30% to more than 40%. AREAS COVERED IN THIS REVIEW Patupilone is a novel cytotoxic compound, with similar microtubule-binding and apoptotic properties of taxanes and is active in taxane-resistant cell lines. Over 1200 patients have been treated with patupilone in Phase I - III clinical trials and a wealth of knowledge has accumulated about this compound. This review discusses current pharmacology and data from clinical trials with patupilone from the last seven years. WHAT THE READER WILL GAIN We present a comprehensive summary of data from Phase II and III trials, as well as an overview of currently accruing trials. TAKE HOME MESSAGE Although patupilone has not demonstrated superiority over pegylated liposomal doxorubicin in a large Phase III trial in relapsed or refractory ovarian cancer, its evaluation is continuing in a range of other malignancies, especially in primary or secondary tumors of the CNS.
Collapse
Affiliation(s)
- Branislav Bystricky
- Department of Medicine, Royal Marsden Hospital, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | | |
Collapse
|
34
|
Zhang K, Heidrich FM, DeGray B, Boehmerle W, Ehrlich BE. Paclitaxel accelerates spontaneous calcium oscillations in cardiomyocytes by interacting with NCS-1 and the InsP3R. J Mol Cell Cardiol 2010; 49:829-35. [PMID: 20801127 DOI: 10.1016/j.yjmcc.2010.08.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2010] [Revised: 08/12/2010] [Accepted: 08/18/2010] [Indexed: 01/09/2023]
Abstract
Paclitaxel (Taxol) is a microtubule-stabilizing compound that is used for cancer chemotherapy. However, Taxol administration is limited by serious side effects including cardiac arrhythmia, which cannot be explained by its microtubule-stabilizing effect. Recently, neuronal calcium sensor 1 (NCS-1), a calcium binding protein that modulates the inositol-1,4,5-trisphosphate receptor (InsP(3)R), was described as a binding partner of Taxol and as a substrate of calpain. We examined calcium signaling processes in cardiomyocytes after treatment with Taxol to investigate the basis of Taxol-induced cardiac arrhythmia. After treating isolated neonatal rat ventricular myocytes with a therapeutic concentration of Taxol for several hours live cell imaging experiments showed that the frequency of spontaneous calcium oscillations significantly increased. This effect was not mimicked by other tubulin-stabilizing agents. However, it was prevented by inhibiting the InsP(3)R. Taxol treated cells had increased expression of NCS-1, an effect also detectable after Taxol administration in vivo. Short hairpin RNA mediated knockdown of NCS-1 decreased InsP(3)R dependent intracellular calcium release, whereas Taxol treatment, that increased NCS-1 levels, increased InsP(3)R dependent calcium release. The effects of Taxol were ryanodine receptor independent. At the single channel level Taxol and NCS-1 mediated an increase in InsP(3)R activity. Calpain activity was not affected by Taxol in cardiomyocytes suggesting a calpain independent signaling pathway. In short, our study shows that Taxol impacts calcium signaling and calcium oscillations in cardiomyocytes through NCS-1 and the InsP(3)R.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA
| | | | | | | | | |
Collapse
|
35
|
Thomas SL, Zhao J, Li Z, Lou B, Du Y, Purcell J, Snyder JP, Khuri FR, Liotta D, Fu H. Activation of the p38 pathway by a novel monoketone curcumin analog, EF24, suggests a potential combination strategy. Biochem Pharmacol 2010; 80:1309-16. [PMID: 20615389 DOI: 10.1016/j.bcp.2010.06.048] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 06/26/2010] [Accepted: 06/28/2010] [Indexed: 02/01/2023]
Abstract
Increasing attention has been given to the anticancer effects of curcumin and the ability of this natural product to inhibit cancer cell proliferation. New curcumin analogs have been developed to optimize the in vitro and in vivo activity of the parent compound yet retain the same safety profile. EF24, a fluorinated synthetic analog, surpasses curcumin in its ability to inhibit cancer cell viability and down-regulate TNFα-induced NF-κB activation. Here we report a critical role of the p38-mediated signaling pathway in the determination of lung cancer cell's sensitivity to EF24. We have found that EF24-induced decease of lung cancer cell viability was accompanied by upregulated mitogen-activated protein kinases (MAPK) as evidenced by increased phosphorylation of ERK1/2, JNK, and p38. Pharmacological investigation led to our suggestion that EF24 triggers a negative feedback loop through p38 activation. In support of this model, inhibition of p38, either by small molecule inhibitors or through an RNAi-mediated knockdown approach, enhanced the EF24-induced apoptotic death of A549 cells. Thus, inhibition of p38 may boost the EF24 anticancer effect. Indeed, a combination of EF24 and SB203580, a p38 inhibitor, synergistically inhibited clonogenic activity of A549 lung cancer cells and induced their apoptosis as reflected by poly(ADP-ribose) polymerase cleavage, the accumulation of the sub-G(1) fraction of cells, and apoptotic cell staining. These studies offer a novel strategy that combines the curcumin analog EF24 with a p38 inhibitor for potentially enhanced therapy in the treatment of lung cancer.
Collapse
|
36
|
Zhou T, Bao Y, Ye S, Weng D, Chen G, Lu Y, Ma D, Wang S. Effect of spindle checkpoint on Akt2-mediated paclitaxel-resistance in A2780 ovarian cancer cells. ACTA ACUST UNITED AC 2010; 30:206-11. [PMID: 20407875 DOI: 10.1007/s11596-010-0215-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Indexed: 12/21/2022]
Abstract
Recent evidence has suggested that Akt2 plays an important role in the protection of cells from paclitaxel (PTX)-induced apoptosis and control of the cell cycle. In addition, some scholars suggested that the PTX sensitivity depends on a functional spindle assembly checkpoint. In the present study, we investigated the role of the Akt2/Bub1 cross-talking in apoptosis and cell cycle after exposure of the A2780 ovarian cancer cells to paclitaxel (PTX). Recombinant expression plasmid WT-Akt2 was transfected into A2780 cells by lipofectamine2000, and then the expression level of Akt2 gene was detected by using RT-PCR and Western blotting. Cell apoptosis and cell cycle were detected by flow cytometry and Hoechst 33342 staining after treatment with PTX. Moreover, we compared the expression level of Bub1 in different groups by Western blotting. Our study showed that up-regulation of Akt2 contributed to A2780 ovarian cancer cells overriding PTX-induced G(2)/M arrest, and inhibited Bub1 expression. Our findings might shed light on the molecular mechanism of PTX-induced resistance in ovarian cancer and help develop novel anti-neoplastic strategies.
Collapse
Affiliation(s)
- Ting Zhou
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Xiao D, Singh SV. Phenethyl isothiocyanate sensitizes androgen-independent human prostate cancer cells to docetaxel-induced apoptosis in vitro and in vivo. Pharm Res 2010; 27:722-31. [PMID: 20182772 DOI: 10.1007/s11095-010-0079-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 02/02/2010] [Indexed: 11/25/2022]
Abstract
PURPOSE The present study was undertaken to determine efficacy of phenethyl isothiocyanate (PEITC) for sensitization of androgen-independent human prostate cancer cells (AIPC) to Docetaxel-induced apoptosis using cellular and xenograft models. METHODS Cell viability was determined by trypan blue dye exclusion assay. Microscopy and DNA fragmentation assay were performed to quantify apoptotic cell death in cultured cells. Protein levels were determined by immunoblotting. PC-3 prostate cancer xenograft model was utilized to determine in vivo efficacy of the PEITC and/or Docetaxel treatments. RESULTS Pharmacologic concentrations of PEITC augmented Docetaxel-induced apoptosis in PC-3 and DU145 cells in association with suppression of Bcl-2 and XIAP protein levels and induction of Bax and Bak. The PEITC-Docetaxel combination was markedly more efficacious against PC-3 xenograft in vivo compared with PEITC or Docetaxel alone. Significantly higher counts of apoptotic bodies were also observed in tumor sections from mice treated with the PEITC-Docetaxel combination compared with PEITC or Docetaxel alone. The PEITC and/or Docetaxel-mediated changes in the levels of apoptosis regulating proteins in the tumor were generally consistent with the molecular alterations observed in cultured cells. CONCLUSION These results offer obligatory impetus to test PEITC-Docetaxel combination for the treatment of AIPC in a clinical setting.
Collapse
Affiliation(s)
- Dong Xiao
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
38
|
Stepien A, Grzanka A, Grzanka D, Andrzej Szczepanski M, Helmin-Basa A, Gackowska L. Taxol-induced polyploidy and cell death in CHO AA8 cells. Acta Histochem 2010; 112:62-71. [PMID: 19004483 DOI: 10.1016/j.acthis.2008.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2008] [Revised: 08/18/2008] [Accepted: 09/03/2008] [Indexed: 10/21/2022]
Abstract
The purpose of this study was to assess whether Taxol-induced changes in microtubular dynamics are accompanied by apoptosis. CHO AA8 cells were treated with different Taxol concentrations (0.25microM, 0.5microM, 1microM) for 24h. The effects of Taxol exposure were analyzed using fluorescence microscopy and flow cytometry (TUNEL and annexin V-FITC/propidium iodide assays). 0.25microM Taxol caused the appearance of few multinucleated giant cells exhibiting extensive arrays of fine filaments. Slight increases in the level of polyploidy, phosphatidylserine externalization and in the percentage of TUNEL positive cells were noticed. Concentrations of 0.5 and 1microM resulted in the appearance of a large number of giant cells, which exhibited, depending on the cell, an extensive microtubular network or loose or tightly packed bundles of microtubules. Cells of reduced volume and showing chromatin condensation were also seen. Cell cycle analysis revealed that almost half of the cell population was polyploid. Except in cells exposed to 1microM Taxol, annexin V-FITC/PI labelling did not reveal the loss of plasma membrane integrity or increase in phosphatidylserine externalization; however, TUNEL assay revealed a significant increase in the percentage of cells with DNA fragmentation. These data indicate that CHO AA8 cells treated with Taxol undergo cell death of a type which considerably differs from apoptosis.
Collapse
|
39
|
Monaghan K, Khong T, Smith G, Spencer A. CYT997 causes apoptosis in human multiple myeloma. Invest New Drugs 2009; 29:232-8. [PMID: 19907921 DOI: 10.1007/s10637-009-9350-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 10/27/2009] [Indexed: 11/27/2022]
Abstract
Multiple Myeloma (MM) is an incurable malignancy of mature plasma cells. Microtubule targeting agents (MTAs) are an established class of drug that include many conventional and some novel compounds. MTAs function by inhibiting the polymerisation or depolymerisation of microtubules (MTs) within the cell, disrupting various important cellular functions. We have investigated pre-clinically the novel tubulin polymerisation inhibitor CYT997 for the potential treatment of MM. Here we demonstrate the promising anti-myeloma activity of CYT997 as evidenced by tubulin disruption, inhibition of growth and proliferation, cell cycle arrest and most importantly apoptosis of both human myeloma cell lines (HMCLs) and primary MM cells using nanomolar drug concentrations. CYT997 also synergises with bortezomib to produce more potent anti-MM activity. These in vitro observations were validated in vivo by the ability of CYT997 to significantly prolong survival in a murine model of aggressive systemic myelomatosis. These findings provide a basis for continuing pre-clinical and clinical investigations into the anti-MM effects of CYT997.
Collapse
Affiliation(s)
- Katherine Monaghan
- Myeloma Research Group, Malignant Haematology and Stem Cell Transplantation, South Block, Alfred Hospital, Prahran, Victoria, Australia
| | | | | | | |
Collapse
|
40
|
Huang HC, Shi J, Orth JD, Mitchison TJ. Evidence that mitotic exit is a better cancer therapeutic target than spindle assembly. Cancer Cell 2009; 16:347-58. [PMID: 19800579 PMCID: PMC2758291 DOI: 10.1016/j.ccr.2009.08.020] [Citation(s) in RCA: 241] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 07/11/2009] [Accepted: 08/24/2009] [Indexed: 12/11/2022]
Abstract
Current antimitotics work by perturbing spindle assembly, which activates the spindle assembly checkpoint, causes mitotic arrest, and triggers apoptosis. Cancer cells can resist such killing by premature exit, before cells initiate apoptosis, due to a weak checkpoint or rapid slippage. We reasoned blocking mitotic exit downstream of the checkpoint might circumvent this resistance. Using single-cell approaches, we showed that blocking mitotic exit by Cdc20 knockdown slowed cyclin B1 proteolysis, thus allowed more time for death initiation. Killing by Cdc20 knockdown did not require checkpoint activity and can occur by intrinsic apoptosis or an alternative death pathway when Bcl2 was overexpressed. We conclude targeting Cdc20, or otherwise blocking mitotic exit, may be a better cancer therapeutic strategy than perturbing spindle assembly.
Collapse
Affiliation(s)
- Hsiao-Chun Huang
- Department of Systems Biology, Harvard Medical School, Boston, MA 02215, USA.
| | | | | | | |
Collapse
|
41
|
Michaud LB. The epothilones: how pharmacology relates to clinical utility. Ann Pharmacother 2009; 43:1294-309. [PMID: 19584389 DOI: 10.1345/aph.1m005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To review the pharmacologic properties of a novel class of chemotherapeutic agents, the epothilones, and to summarize findings from recent clinical trials investigating the various epothilones in cancer therapy. DATA SOURCES Literature searches were conducted using MEDLINE, PubMed, and the abstract search engines for the American Society of Clinical Oncology and American Association for Cancer Research annual meetings (all searches through November 2008). Primary search terms included epothilone, BMS-247550, ixabepilone, EPO906, patupilone, sagopilone, and ZK-EPO. STUDY SELECTION AND DATA EXTRACTION Publications were given priority for inclusion if they discussed structural or pharmacologic properties of the epothilones as a class or if they included preclinical or clinical data for epothilones currently in clinical development. DATA SYNTHESIS The epothilones are a novel class of microtubule-stabilizing agents (MSAs). Epothilones are structurally and pharmacologically distinct from taxanes, but the exact ways in which the pharmacophores of the 2 classes differ has not been firmly established. A number of natural, semisynthetic, and fully synthetic epothilones are in various stages of clinical development. These agents differ from each other and from existing MSAs; these differences influence potency, stability, and solubility. Ixabepilone is currently approved to treat multidrug-resistant metastatic breast cancer and has demonstrated efficacy in earlier stages of breast cancer and in several other tumor types. Patupilone and sagopilone are currently under clinical investigation and have each shown promise in a number of treatment settings and tumor types. All 3 agents appear to be associated with manageable toxicities, but no class-wide toxicity profile exists for the epothilones and dose-limiting toxicities differ among the agents. CONCLUSIONS The epothilones have demonstrated significant potential for addressing the growing therapeutic challenge of taxane resistance, and the ever-increasing pool of information regarding structure-activity relationships of these MSAs will help to optimize microtubule-targeted chemotherapy.
Collapse
Affiliation(s)
- Laura Boehnke Michaud
- Clinical Pharmacy Services, Division of Pharmacy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
42
|
Zhao Y, Fang WS, Pors K. Microtubule stabilising agents for cancer chemotherapy. Expert Opin Ther Pat 2009; 19:607-22. [DOI: 10.1517/13543770902775713] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
43
|
Salum L, Dias L, Andricopulo A. Fragment-Based QSAR and Molecular Modeling Studies on a Series of Discodermolide Analogs as Microtubule-Stabilizing Anticancer Agents. ACTA ACUST UNITED AC 2009. [DOI: 10.1002/qsar.200860109] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
44
|
Beyer CF, Zhang N, Hernandez R, Vitale D, Nguyen T, Ayral-Kaloustian S, Gibbons JJ. The microtubule-active antitumor compound TTI-237 has both paclitaxel-like and vincristine-like properties. Cancer Chemother Pharmacol 2009; 64:681-9. [PMID: 19132373 DOI: 10.1007/s00280-008-0916-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Accepted: 12/21/2008] [Indexed: 02/04/2023]
Abstract
PURPOSE To compare TTI-237 (5-chloro-6-[2,6-difluoro-4-[3-(methylamino)propoxy]phenyl]-N-[(1S)-2,2,2-trifluoro-1-methylethyl]-[1, 2, 4]triazolo[1,5-a]pyrimidin-7-amine butanedioate) with paclitaxel and vincristine in order to better understand the properties of this new anti-microtubule agent. METHODS Tubulin polymerization and depolymerization were followed by turbidimetric assays. Effects of compounds on the binding of [(3)H]guanosine triphosphate ([(3)H]GTP) to tubulin were studied by competition binding assays. Effects of compounds on the phosphorylation of a panel of intracellular proteins were determined by flow cytometry using phosphoprotein-specific antibodies. RESULTS At low molar ratios of TTI-237:tubulin heterodimer (about 1:30), TTI-237 enhanced depolymerization kinetics in response to low temperature, but stabilized the aggregates at higher ratios (about 1:4). Similarly, the aggregates induced in microtubule protein by TTI-237 were depolymerized by excess Ca(++) at low TTI-237:tubulin-heterodimer molar ratios, but were stable at higher ratios. TTI-237 inhibited the exchange of [(3)H]GTP at the exchangeable nucleotide site of the tubulin heterodimer, and was similar to vincristine in its effects on the phosphorylation of eight intracellular proteins in HeLa cells. CONCLUSIONS TTI-237 has properties that distinguish it from typical vinca-site and taxoid-site ligands, and therefore it may exemplify a new class of microtubule-active compounds.
Collapse
Affiliation(s)
- Carl F Beyer
- Department of Discovery Oncology, Wyeth Research, Pearl River, NY 10965, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Araki H, Katoh T. Total Synthesis of Otteliones Possessing Powerful Tubulin Polymerization Inhibitory Activity. J SYN ORG CHEM JPN 2009. [DOI: 10.5059/yukigoseikyokaishi.67.909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
46
|
Total synthesis of novel dictyostatin analogs and hybrids as microtubule-stabilizing anticancer agents. PURE APPL CHEM 2009. [DOI: 10.1351/pac-con-08-09-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Structural modification of the dictyostatin macrolide template through adaptation of our total synthesis has led to the identification of a number of potent analogs of this novel microtubule-stabilizing agent. A common synthetic strategy was exploited, employing a (Z)-selective Still-Gennari olefination between various advanced C11-C26 aldehyde and C4-C10 (or C1-C10) β-ketophosphonate intermediates. In vitro evaluation of the growth inhibitory activity of these analogs against both Taxol-sensitive and -resistant human cancer cell lines has provided a foundation for structure-activity relationship (SAR) studies to help define the pharmacophore region.
Collapse
|
47
|
Autotaxin protects MCF-7 breast cancer and MDA-MB-435 melanoma cells against Taxol-induced apoptosis. Oncogene 2008; 28:1028-39. [PMID: 19079345 DOI: 10.1038/onc.2008.442] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Autotaxin (ATX) promotes cancer cell survival, growth, migration, invasion and metastasis. ATX converts extracellular lysophosphatidylcholine (LPC) into lysophosphatidate (LPA). As these lipids have been reported to affect cell signaling through their own G-protein-coupled receptors, ATX could modify the balance of this signaling. Also, ATX affects cell adhesion independently of its catalytic activity. We investigated the interactions of ATX, LPC and LPA on the apoptotic effects of Taxol, which is commonly used in breast cancer treatment. LPC had no significant effect on Taxol-induced apoptosis in MCF-7 breast cancer cells, which do not secrete significant ATX. Addition of incubation medium from MDA-MB-435 melanoma cells, which secrete ATX, or recombinat ATX enabled LPC to inhibit Taxol-induced apoptosis of MCF-7 cells. Inhibiting ATX activity blocked this protection against apoptosis. We conclude that LPC has no significant effect in protecting MCF-7 cells against Taxol treatment unless it is converted to LPA by ATX. LPA strongly antagonized Taxol-induced apoptosis through stimulating phosphatidylinositol 3-kinase and inhibiting ceramide formation. LPA also partially reversed the Taxol-induced arrest in the G2/M phase of the cell cycle. Our results support the hypothesis that therapeutic inhibition of ATX activity could improve the efficacy of Taxol as a chemotherapeutic agent for cancer treatment.
Collapse
|
48
|
Epothilones: tubulin polymerization as a novel target for prostate cancer therapy. ACTA ACUST UNITED AC 2008; 6:85-92. [DOI: 10.1038/ncponc1281] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2007] [Accepted: 04/22/2008] [Indexed: 01/05/2023]
|
49
|
Paterson I, Gardner NM, Guzmán E, Wright AE. Total synthesis and biological evaluation of novel C2-C6 region analogues of dictyostatin. Bioorg Med Chem 2008; 17:2282-9. [PMID: 19022679 DOI: 10.1016/j.bmc.2008.10.084] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Revised: 06/12/2008] [Accepted: 10/31/2008] [Indexed: 11/26/2022]
Abstract
By exploiting a Still-Gennari HWE coupling with a common C11-C26 aldehyde, a series of C2-C6 modified analogues of the microtubule-stabilising marine natural product dictyostatin were synthesised and evaluated in vitro for growth inhibition against a range of human cancer cell lines, including the (P-glycoprotein efflux-mediated) Taxol-resistant NCI/ADR cell line. Removal of the C6 methyl substituent in dictyostatin was found to be well tolerated and led to the retention of antiproliferative activity in the low nanomolar range (IC(50)=43 nM in the NCI/ADR cell line), while partial and full saturation of the (2Z,4E)-dienoate region led to a progressive reduction in biological potency. The lactone ring size was found to be critical, as C21 to C19 translactonisation to afford 20-membered isodictyostatin analogues led to a significant loss of cytotoxicity. In a series of incubatory experiments performed on the PANC-1 cell line, all three of the 22-membered macrolide analogues acted in an analogous fashion to dictyostatin, through a mechanism of microtubule stabilization, causing both an accumulation of cells at the G2/M phase and formation of characteristic dense intracellular microtubule bundles.
Collapse
Affiliation(s)
- Ian Paterson
- University Chemical Laboratory, Lensfield Road, Cambridge CB2 1EW, UK.
| | | | | | | |
Collapse
|
50
|
Paterson I, Gardner NM, Guzmán E, Wright AE. Total synthesis and biological evaluation of potent analogues of dictyostatin: modification of the C2-C6 dienoate region. Bioorg Med Chem Lett 2008; 18:6268-72. [PMID: 18951787 DOI: 10.1016/j.bmcl.2008.09.109] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Accepted: 09/22/2008] [Indexed: 11/30/2022]
Abstract
By exploiting a Still-Gennari olefination of a common C11-C26 aldehyde with a C4-C10 or C1-C10 beta-ketophosphonate, three modified C2-C6 region analogues of the 22-membered macrolide dictyostatin were synthesised and evaluated in vitro for growth inhibition against a range of human cancer cell lines, including the Taxol-resistant NCI/ADR-Res cell line. 6-Desmethyldictyostatin and 2,3-dihydrodictyostatin displayed potent (low nanomolar) antiproliferative activity, intermediate between dictyostatin and discodermolide, while 2,3,4,5-tetrahydrodictyostatin showed activity comparable to discodermolide. As with dictyostatin, these simplified analogues act through a mechanism of microtubule stabilisation, G2/M arrest and apoptosis.
Collapse
Affiliation(s)
- Ian Paterson
- University Chemical Laboratory, Lensfield Road, Cambridge CB2 1EW, UK.
| | | | | | | |
Collapse
|