1
|
Liu X, Hao Z, He H, Wang X, Wang W, Shu X, Sun B, Hu Z, Hu S, Hou X, Xiao Y, Zhou H, Liu Y, Wang J, Fu Z. Accumulation of microtubule-associated protein tau promotes hepatocellular carcinogenesis through inhibiting autophagosome-lysosome fusion. Mol Cell Biochem 2025; 480:3621-3635. [PMID: 39718681 DOI: 10.1007/s11010-024-05193-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Dysregulated expression of microtubule-associated protein tau (MAPT) has been reported in a variety of human cancers. However, whether and how Tau influences hepatocellular carcinogenesis remains elusive. This study was aimed to investigate the role and the underlying mechanism of Tau in the proliferation, invasion, migration and sorafenib sensitivity of hepatocellular carcinoma (HCC) cells. An increased level of Tau was found in the primary tumor samples of HCC compared with the adjacent normal liver tissues, and the increase of Tau was positively correlated with p62 evidenced by the data obtained from The Cancer Genome Atlas (TCGA), Gene Expression Profiling Interactive Analysis (GEPIA), and human samples from HCC patients. The high Tau expression was also correlated with a poorer survival in HCC patients demonstrated by using the GEPIA survival analysis and OncoLnc database. Further studies showed that Tau overexpression promoted the growth, invasion and migration and decreased sorafenib sensitivity in HepG2 and Huh7 cells; Tau also accelerated growth of xenograft tumors with blockage of autophagosome-lysosome fusion. Finally, overexpressing Tau inhibited AMPK, which contributed to Tau-induced promotion of hepatocellular carcinogenesis. In conclusion, our study provides the proof-of-concept evidence validating Tau as an attractive HCC target.
Collapse
Affiliation(s)
- Xuemin Liu
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, 8 Sanjiaohu Road, Wuhan, 430056, China
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Zhiwei Hao
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, 8 Sanjiaohu Road, Wuhan, 430056, China
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Huanhuan He
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, 8 Sanjiaohu Road, Wuhan, 430056, China
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Xuan Wang
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, 8 Sanjiaohu Road, Wuhan, 430056, China
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Wenqi Wang
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, 8 Sanjiaohu Road, Wuhan, 430056, China
| | - Xiji Shu
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, 8 Sanjiaohu Road, Wuhan, 430056, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, 430056, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Binlian Sun
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, 430056, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Zhiyong Hu
- Department of Pathology, Renmin Hospital of Huangpi District of Jianghan University, Wuhan, 430399, China
| | - Shaobo Hu
- Liver Transplant Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoying Hou
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Yue Xiao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongyan Zhou
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, 8 Sanjiaohu Road, Wuhan, 430056, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, 430056, China
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Yuchen Liu
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China.
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, 430056, China.
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.
| | - Jianzhi Wang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan, 430056, China.
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.
| | - Zhengqi Fu
- Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, 8 Sanjiaohu Road, Wuhan, 430056, China.
- Cancer Institute, School of Medicine, Jianghan University, Wuhan, 430056, China.
| |
Collapse
|
2
|
Pan Q, Luo P, Qiu Y, Hu K, Lin L, Zhang H, Yin D, Shi C. The SETDB1-PC4-UPF1 post-transcriptional machinery controls periodic degradation of CENPF mRNA and maintains mitotic progression. Cell Death Differ 2025:10.1038/s41418-025-01465-z. [PMID: 40016337 DOI: 10.1038/s41418-025-01465-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 01/31/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
Numerous genes exhibit periodic oscillations in mRNA expression, essential for orderly cell division. Mitosis-related mRNAs fluctuate cyclically from the G2 to M phase, primarily regulated by transcription factors. However, the role of post-transcriptional regulation in this process remains unclear. Here, we demonstrated a decrease in mRNA levels of centromere protein F (CENPF) from the early to late G2 phase. SETDB1-PC4-UPF1 serves as a crucial post-transcriptional machinery, orchestrating the periodic degradation of CENPF mRNA, ensuring balanced CENP expression, proper spindle assembly, and successful mitosis. In early G2, newly synthesized CENPF mRNAs accumulate and bind to PC4, leading to SETDB1-mediated PC4 dimethylation at K35. In late G2, dimethylated PC4 interacts with UPF1 to promote deadenylation-dependent degradation of CENPF mRNAs, forming a regulatory loop for CENP homeostasis. Elevated PC4 dimethylation in hepatocellular carcinoma, coupled with increased sensitivity to taxanes upon its inhibition, suggests promising therapeutic avenues. These findings suggest a post-transcriptional quality control mechanism regulating cyclic mitotic mRNA fluctuations, providing comprehensive insights into cell cycle gene regulation dynamics.
Collapse
Affiliation(s)
- Qimei Pan
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Peng Luo
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China
| | - Yuntan Qiu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kaishun Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lehang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Heyun Zhang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China.
| |
Collapse
|
3
|
Barker RM, Chambers A, Kehoe PG, Rowe E, Perks CM. Untangling the role of tau in sex hormone responsive cancers: lessons learnt from Alzheimer's disease. Clin Sci (Lond) 2024; 138:1357-1369. [PMID: 39469929 PMCID: PMC11522895 DOI: 10.1042/cs20230317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/20/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Tubulin associated unit has been extensively studied in neurodegenerative diseases including Alzheimer's disease (AD), whereby its hyperphosphorylation and accumulation contributes to disease pathogenesis. Tau is abundantly expressed in the central nervous system but is also present in non-neuronal tissues and in tumours including sex hormone responsive cancers such as breast and prostate. Curiously, hormonal effects on tau also exist in an AD context from numerous studies on menopause, hormone replacement therapy, and androgen deprivation therapy. Despite sharing some risk factors, most importantly advancing age, there are numerous reports from population studies of, currently poorly explained inverse associations between cancer and Alzheimer's disease. We previously reviewed important components of the phosphoinositide-3-kinase/protein kinase B (PI3K/Akt) signalling pathway and their differential modulation in relation to the two diseases. Similarly, receptor tyrosine kinases, estrogen receptor and androgen receptor have all been implicated in the pathogenesis of both cancer and AD. In this review, we focus on tau and its effects in hormone responsive cancer in terms of development, progression, and treatment and in relation to sex hormones and PI3K/Akt signalling molecules including IRS-1, PTEN, Pin1, and p53.
Collapse
Affiliation(s)
- Rachel M. Barker
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Alfie Chambers
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Patrick G. Kehoe
- Department of Urology, Bristol Urological Institute, Southmead Hospital, Bristol BS10 5NB, UK
| | - Edward Rowe
- Dementia Research Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Claire M. Perks
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| |
Collapse
|
4
|
Firdous F, Riaz S, Furqan M, Fozail S, Fatima K, Pohl SÖG, Doleschall NJ, Myant KB, Kahfi J, Emwas AH, Jaremko M, Chotana GA, Saleem RSZ, Faisal A. Design, Synthesis, and Biological Evaluation of SSE1806, a Microtubule Destabilizer That Overcomes Multidrug Resistance. ACS Med Chem Lett 2023; 14:1369-1377. [PMID: 37849542 PMCID: PMC10577696 DOI: 10.1021/acsmedchemlett.3c00258] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/06/2023] [Indexed: 10/19/2023] Open
Abstract
Microtubules are dynamic structures that form spindle fibers during cell division; pharmacological inhibition of microtubule dynamics arrests cells in mitosis, leading to apoptosis, and they have been extensively used to treat various cancers. However, the efficacy of such drugs is often limited by multidrug resistance. This study synthesized and evaluated 30 novel derivatives of podophyllotoxin, a natural antimitotic compound, for their antiproliferative activities. Compound SSE1806 exhibited the most potent antiproliferative activity with GI50 values ranging from 1.29 ± 0.01 to 21.15 ± 2.1 μM in cancer cell lines of different origins; it directly inhibited microtubule polymerization, causing aberrant mitosis and G2/M arrest. Prolonged treatment with SSE1806 increased p53 expression, induced cell death in monolayer cultures, and reduced the growth of mouse- and patient-derived human colon cancer organoids. Importantly, SSE1806 overcame multidrug resistance in a cell line overexpressing MDR-1. Thus, SSE1806 represents a potential anticancer agent that can overcome multidrug resistance.
Collapse
Affiliation(s)
- Farhat Firdous
- Department
of Chemistry and Chemical Engineering, Syed Babar Ali School of Science
and Engineering, Lahore University of Management
Sciences, Lahore 54792, Pakistan
- Department
of Life Sciences, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore 54792, Pakistan
| | - Sharon Riaz
- Department
of Chemistry and Chemical Engineering, Syed Babar Ali School of Science
and Engineering, Lahore University of Management
Sciences, Lahore 54792, Pakistan
| | - Muhammad Furqan
- Department
of Life Sciences, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore 54792, Pakistan
| | - Salman Fozail
- Department
of Life Sciences, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore 54792, Pakistan
| | - Khushboo Fatima
- Department
of Life Sciences, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore 54792, Pakistan
| | - Sebastian Öther-Gee Pohl
- Institute
of Genetics and Cancer, The University of
Edinburgh, Western General Hospital Campus, Crewe Road, Edinburgh EH4 2XU, Scotland
| | - Nora Julia Doleschall
- Institute
of Genetics and Cancer, The University of
Edinburgh, Western General Hospital Campus, Crewe Road, Edinburgh EH4 2XU, Scotland
| | - Kevin B. Myant
- Institute
of Genetics and Cancer, The University of
Edinburgh, Western General Hospital Campus, Crewe Road, Edinburgh EH4 2XU, Scotland
| | - Jordan Kahfi
- Division
of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Abdul-Hamid Emwas
- KAUST
Core Laboratories, King Abdullah University
of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Mariusz Jaremko
- Division
of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Ghayoor Abbas Chotana
- Department
of Chemistry and Chemical Engineering, Syed Babar Ali School of Science
and Engineering, Lahore University of Management
Sciences, Lahore 54792, Pakistan
| | - Rahman Shah Zaib Saleem
- Department
of Chemistry and Chemical Engineering, Syed Babar Ali School of Science
and Engineering, Lahore University of Management
Sciences, Lahore 54792, Pakistan
| | - Amir Faisal
- Department
of Life Sciences, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore 54792, Pakistan
| |
Collapse
|
5
|
Ahn S, Kwon A, Oh Y, Rhee S, Song WK. Microtubule Acetylation-Specific Inhibitors Induce Cell Death and Mitotic Arrest via JNK/AP-1 Activation in Triple-Negative Breast Cancer Cells. Mol Cells 2023; 46:387-398. [PMID: 36794420 PMCID: PMC10258459 DOI: 10.14348/molcells.2023.2192] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 02/17/2023] Open
Abstract
Microtubule acetylation has been proposed as a marker of highly heterogeneous and aggressive triple-negative breast cancer (TNBC). The novel microtubule acetylation inhibitors GM-90257 and GM-90631 (GM compounds) cause TNBC cancer cell death but the underlying mechanisms are currently unknown. In this study, we demonstrated that GM compounds function as anti-TNBC agents through activation of the JNK/AP-1 pathway. RNA-seq and biochemical analyses of GM compound-treated cells revealed that c-Jun N-terminal kinase (JNK) and members of its downstream signaling pathway are potential targets for GM compounds. Mechanistically, JNK activation by GM compounds induced an increase in c-Jun phosphorylation and c-Fos protein levels, thereby activating the activator protein-1 (AP-1) transcription factor. Notably, direct suppression of JNK with a pharmacological inhibitor alleviated Bcl2 reduction and cell death caused by GM compounds. TNBC cell death and mitotic arrest were induced by GM compounds through AP-1 activation in vitro. These results were reproduced in vivo, validating the significance of microtubule acetylation/JNK/AP-1 axis activation in the anti-cancer activity of GM compounds. Moreover, GM compounds significantly attenuated tumor growth, metastasis, and cancer-related death in mice, demonstrating strong potential as therapeutic agents for TNBC.
Collapse
Affiliation(s)
- Suyeon Ahn
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| | - Ahreum Kwon
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| | - Youngsoo Oh
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Woo Keun Song
- Cell Logistics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| |
Collapse
|
6
|
Zhang J, Tan L, Wu C, Li Y, Chen H, Liu Y, Wang Y. Discovery and biological evaluation of 4,6-pyrimidine analogues with potential anticancer agents as novel colchicine binding site inhibitors. Eur J Med Chem 2023; 248:115085. [PMID: 36621138 DOI: 10.1016/j.ejmech.2022.115085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Novel 4,6-pyrimidine analogues were designed and synthesized as colchicine binding site inhibitors (CBSIs) with potent antiproliferative activities. Among them, compound 17j has the most potent activities against 6 human cancer cell lines with IC50 values from 1.1 nM to 4.4 nM, which was 76 times higher than the lead compound 3 in A549 cells. The co-crystal structure of 17j in complex with tubulin confirms the key binding mode at the colchicine binding site. Moreover, 17j inhibited the tubulin polymerization in biochemical assays, depolymerized cellular microtubules, induced the G2/M arrest, inhibited the cell migration, and promoted the initiation of apoptosis. In vivo, 17j effectively inhibits primary tumor growth with tumor growth inhibition rates of 42.51% (5 mg/kg) and 65.42% (10 mg/kg) in A549 xenograft model. Taken together, 17j represents a promising new generation of CBSIs.
Collapse
Affiliation(s)
- Jifa Zhang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Tianfu Jincheng Laboratory, Chengdu, 610041, Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lun Tan
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chengyong Wu
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Tianfu Jincheng Laboratory, Chengdu, 610041, Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuyan Li
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Yinghuan Liu
- Tianfu Jincheng Laboratory, Chengdu, 610041, Sichuan, China
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Tianfu Jincheng Laboratory, Chengdu, 610041, Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
7
|
Kumari A, Prassanawar SS, Panda D. β-III Tubulin Levels Determine the Neurotoxicity Induced by Colchicine-Site Binding Agent Indibulin. ACS Chem Neurosci 2023; 14:19-34. [PMID: 36541944 DOI: 10.1021/acschemneuro.2c00324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Indibulin, a microtubule-depolymerizing agent, produces minimal neurotoxicity in animals. It is also less cytotoxic toward differentiated neuronal cells than undifferentiated cells. We found that the levels of β-III tubulin, acetylated tubulin, and polyglutamylated tubulin were significantly increased in differentiated neuroblastoma cells (SH-SY5Y). Since neuronal cells express β-tubulin isotypes differently from other cell types, we explored the binding of indibulin to different β-tubulin isotypes. Our molecular docking analysis suggested that indibulin binds to β-III tubulin with lower affinity than to other β-tubulin isotypes. We therefore studied the implications of different β-tubulin isotypes on the cytotoxic effects of indibulin, colchicine, and vinblastine in differentiated SH-SY5Y cells. Upon depletion of β-III tubulin in the differentiated cells, the toxicity of indibulin and colchicine significantly increased, while sensitivity to vinblastine was unaffected. Using biochemical, bioinformatics, and fluorescence spectroscopic techniques, we have identified the binding site of indibulin on tubulin, which had not previously been established. Indibulin inhibited the binding of colchicine and C12 (a colchicine-site binder) to tubulin and also increased the dissociation constant of the interaction between tubulin and colchicine. Indibulin did not inhibit the binding of vinblastine or taxol to tubulin. Interestingly, indibulin antagonized colchicine treatment but synergized with vinblastine treatment in a combination study performed in MDA-MB-231 cells. The results indicate that indibulin is a colchicine-site binder and that the efficacy of colchicine-site binders is affected by the β-III tubulin levels in the cells.
Collapse
Affiliation(s)
- Anuradha Kumari
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Shweta S Prassanawar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India.,National Institute of Pharmaceutical Education and Research, SAS Nagar, Punjab 160062, India
| |
Collapse
|
8
|
Butt NUH, Baytas SN. Advancements in Hepatocellular Carcinoma: Potential Preclinical Drugs and their Future. Curr Pharm Des 2023; 29:2-14. [PMID: 36529919 DOI: 10.2174/1381612829666221216114350] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/12/2022] [Accepted: 10/27/2022] [Indexed: 12/23/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the foremost causes of tumor-affiliated demises globally. The HCC treatment has undergone numerous developments in terms of both drug and non-drug treatments. The United States Food and Drug Administration (FDA) has authorized the usage of a variety of drugs for the treatment of HCC in recent years, involving multi-kinase inhibitors (lenvatinib, regorafenib, ramucirumab, and cabozantinib), immune checkpoint inhibitors (ICIs) (pembrolizumab and nivolumab), and combination therapies like atezolizumab along with bevacizumab. There are currently over a thousand ongoing clinical and preclinical studies for novel HCC drugs, which portrays a competent setting in the field. This review discusses the i. FDA-approved HCC drugs, their molecular targets, safety profiles, and potential disadvantages; ii. The intrial agents/drugs, their molecular targets, and possible benefits compared to alternatives, and iii. The current and future status of potential preclinical drugs with novel therapeutic targets for HCC. Consequently, existing drug treatments and novel strategies with their balanced consumption could ensure a promising future for a universal remedy of HCC in the near future.
Collapse
Affiliation(s)
- Noor-Ul-Huda Butt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkiye
| | - Sultan Nacak Baytas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkiye
| |
Collapse
|
9
|
Matboli M, Hasanin AH, Hussein R, El-Nakeep S, Habib EK, Ellackany R, Saleh LA. Cyanidin 3-glucoside modulated cell cycle progression in liver precancerous lesion, in vivo study. World J Gastroenterol 2021; 27:1435-1450. [PMID: 33911466 PMCID: PMC8047539 DOI: 10.3748/wjg.v27.i14.1435] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/22/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cyanidin-3-O-glucoside (cyan) exhibits antioxidant and anticancer properties. The cell cycle proteins and antimitotic drugs might be promising therapeutic targets in hepatocellular carcinoma. AIM To investigate the effect of cyan administration on cell cycle in hepatic precancerous lesion (PCL) induced by diethylnitrosamine/2-acetylaminofluorene (DEN/2-AAF) in Wistar rats. METHODS In vivo, DEN/2-AAF-induced hepatic PCL, rats were treated with three doses of cyan (10, 15, and 20 mg/kg/d, for four consecutive days per week for 16 wk). Blood and liver tissue samples were collected for measurement of the followings; alpha fetoprotein (AFP) liver function and RNA panel differential expression was evaluated via real time polymerase chain reaction. Histopathological examination of liver sections stained with H&E and immunohistochemical study using glutathione S-transferase placental (GSTP) and proliferating cell nuclear antigen (PCNA) antibodies were assessed. RESULTS Cyan administration mitigated the effect of DEN/2-AFF induced PCL, decreased AFP levels, and improved liver function. Remarkably, treatment with cyan dose dependently decreased the long non-coding RNA MALAT1 and tubulin gamma 1 mRNA expressions and increased the levels of miR-125b, all of which are involved in cell cycle and mitotic spindle assembly. Of note, cyan decreased GSTP foci percent area and PCNA positively stained nuclei. CONCLUSION Our results indicated that cyan could be used as a potential therapeutic agent to inhibit liver carcinogenesis in rat model via modulation of cell cycle.
Collapse
Affiliation(s)
- Marwa Matboli
- Department of Biochemistry, Ain Shams Faculty of Medicine, Cairo 11318, Egypt
| | - Amany H Hasanin
- Department of Clinical Pharmacology, Ain Shams Faculty of Medicine, Cairo 11381, Egypt
| | - Reham Hussein
- Department of Clinical Pharmacology, Ain Shams Faculty of Medicine, Cairo 11381, Egypt
| | - Sarah El-Nakeep
- Department of General Internal Medicine, Ain Shams Faculty of Medicine, Cairo 11381, Egypt
| | - Eman K Habib
- Department of Anatomy & Embryology, Ain Shams Faculty of Medicine, Cairo 11318, Egypt
| | - Rawan Ellackany
- Department of Undergraduate, Faculty of Medicine, Modern University for Technology and Information, Cairo 11381, Egypt
| | - Lobna A Saleh
- Department of Clinical Pharmacology, Ain Shams Faculty of Medicine, Cairo 11381, Egypt
| |
Collapse
|
10
|
Zhong FJ, Sun B, Cao MM, Xu C, Li YM, Yang LY. STMN2 mediates nuclear translocation of Smad2/3 and enhances TGFβ signaling by destabilizing microtubules to promote epithelial-mesenchymal transition in hepatocellular carcinoma. Cancer Lett 2021; 506:128-141. [PMID: 33705863 DOI: 10.1016/j.canlet.2021.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 12/24/2022]
Abstract
Metastasis remains the major obstacle of improving the survival of patients with hepatocellular carcinoma (HCC). Epithelial-mesenchymal transition (EMT) is critical to cancer metastasis. Successful induction of EMT requires dramatic cytoskeleton rearrangement. However, the significance of microtubule (MT), one of the core components of cell cytoskeleton, in this process remains largely unknown. Here we revealed that STMN2, an important MT dynamics regulator, is barely expressed in normal live tissues but markedly up-regulated in HCCs, especially in those with early recurrence. High STMN2 expression correlates with aggressive clinicopathological features and predicts poor prognosis of HCC patients. STMN2 overexpression in HCC cells promotes EMT, invasion and metastasis in vitro and in vivo, whereas STMN2 knockdown has opposite results. Mechanistically, STMN2 modulates MTs disassembly, disrupts MT-Smad complex, and facilitates release from MT network, phosphorylation and nuclear translocation of Smad2/3 even independent of TGFβ stimulation, thereby enhancing TGFβ signaling. Collectively, STMN2 orchestrates MT disassembly to facilitate EMT via TGF-β signaling, providing a novel insight into the mechanisms underlying cancer metastasis. STMN2 is a promising prognostic biomarker and potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Fang-Jing Zhong
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Bo Sun
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Mo-Mo Cao
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Cong Xu
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yi-Ming Li
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lian-Yue Yang
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
11
|
Falgàs A, Pallarès V, Unzueta U, Núñez Y, Sierra J, Gallardo A, Alba-Castellón L, Mangues MA, Álamo P, Villaverde A, Vázquez E, Mangues R, Casanova I. Specific Cytotoxic Effect of an Auristatin Nanoconjugate Towards CXCR4 + Diffuse Large B-Cell Lymphoma Cells. Int J Nanomedicine 2021; 16:1869-1888. [PMID: 33716502 PMCID: PMC7944372 DOI: 10.2147/ijn.s289733] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Around 40-50% of diffuse large-B cell lymphoma (DLBCL) patients suffer from refractory disease or relapse after R-CHOP first-line treatment. Many ongoing clinical trials for DLBCL patients involve microtubule targeting agents (MTAs), however, their anticancer activity is limited by severe side effects. Therefore, we chose to improve the therapeutic window of the MTA monomethyl auristatin E developing a nanoconjugate, T22-AUR, that selectively targets the CXCR4 receptor, which is overexpressed in many DLBCL cells (CXCR4+) and associated with poor prognosis. METHODS The T22-AUR specificity towards CXCR4 receptor was performed by flow cytometry in different DLBCL cell lines and running biodistribution assays in a subcutaneous mouse model bearing CXCR4+ DLBCL cells. Moreover, we determined T22-AUR cytotoxicity using cell viability assays, cell cycle analysis, DAPI staining and immunohistochemistry. Finally, the T22-AUR antineoplastic effect was evaluated in vivo in an extranodal CXCR4+ DLBCL mouse model whereas the toxicity analysis was assessed by histopathology in non-infiltrated mouse organs and by in vitro cytotoxic assays in human PBMCs. RESULTS We demonstrate that the T22-AUR nanoconjugate displays CXCR4-dependent targeting and internalization in CXCR4+ DLBCL cells in vitro as well as in a subcutaneous DLBCL mouse model. Moreover, it shows high cytotoxic effect in CXCR4+ DLBCL cells, including induction of G2/M mitotic arrest, DNA damage, mitotic catastrophe and apoptosis. Furthermore, the nanoconjugate shows a potent reduction in lymphoma mouse dissemination without histopathological alterations in non-DLBCL infiltrated organs. Importantly, T22-AUR also exhibits lack of toxicity in human PBMCs. CONCLUSION T22-AUR exerts in vitro and in vivo anticancer effect on CXCR4+ DLBCL cells without off-target toxicity. Thus, T22-AUR promises to become an effective therapy for CXCR4+ DLBCL patients.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Death/drug effects
- Cell Line, Tumor
- Disease Models, Animal
- Endocytosis/drug effects
- Female
- Humans
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/pathology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lysosomes/drug effects
- Lysosomes/metabolism
- Mice, Inbred NOD
- Mice, SCID
- Nanoconjugates/therapeutic use
- Oligopeptides/pharmacology
- Oligopeptides/therapeutic use
- Receptors, CXCR4/metabolism
- Signal Transduction/drug effects
- Subcutaneous Tissue/drug effects
- Subcutaneous Tissue/pathology
- Tissue Distribution/drug effects
- Mice
Collapse
Affiliation(s)
- Aïda Falgàs
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| | - Victor Pallarès
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| | - Ugutz Unzueta
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, 08193, Spain
| | - Yáiza Núñez
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
| | - Jorge Sierra
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
| | - Alberto Gallardo
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
| | - Lorena Alba-Castellón
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
| | - Maria Antonia Mangues
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
- Department of Pharmacy, Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
| | - Patricia Álamo
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| | - Antonio Villaverde
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, 08193, Spain
- Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, Barcelona, 08193, Spain
| | - Esther Vázquez
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, 08193, Spain
- Institute of Biotechnology and Biomedicine (IBB), Universitat Autònoma de Barcelona, Barcelona, 08193, Spain
| | - Ramon Mangues
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| | - Isolda Casanova
- Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, 08025, Spain
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, 08916, Spain
- CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, 28029, Spain
| |
Collapse
|
12
|
Joshi AM, Prousi GS, Bianco C, Malla M, Guha A, Shah M, Brown SA, Patel B. Microtubule Inhibitors and Cardiotoxicity. Curr Oncol Rep 2021; 23:30. [PMID: 33582937 PMCID: PMC8414965 DOI: 10.1007/s11912-021-01014-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2021] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW Cancer and heart disease are the leading causes of mortality in the USA. Advances in cancer therapies, namely, the development and use of chemotherapeutic agents alone or in combination, are becoming increasingly prevalent. RECENT FINDINGS Many chemotherapeutic agents have been associated with adverse cardiovascular manifestations. The mechanisms of these sequelae remain incompletely understood. In particular, microtubule inhibitor (MTI) agents have been related to the development of heart failure, myocardial ischemia, and conduction abnormalities. At present, there are no guidelines for patients undergoing MTI therapy as it pertains to both preventative and mitigatory strategies for cardiovascular complications. We conducted a literature review focusing on content related to the use of MTIs and their effect on the cardiovascular system. MTIs have been associated with various forms of cardiotoxicity, and fatal cardiotoxicities are rare. The most well-described cardiotoxicities are brady- and tachyarrhythmias. The co-administration of anthracycline-based agents with MTIs can increase the risk of cardiotoxicity.
Collapse
Affiliation(s)
- Amogh M Joshi
- Department of Internal Medicine, Lehigh Valley Health Network, Allentown, PA, USA
| | - George S Prousi
- Department of Cardiology, University of South Carolina, Columbia, SC, USA
| | - Christopher Bianco
- Heart and Vascular Institute, West Virginia University, 1 Medical Center Dr., Morgantown, WV, 26505, USA
| | - Midhun Malla
- Hematology and Oncology, West Virginia University, Morgantown, WV, USA
| | - Avirup Guha
- Department of Cardiology, Case Western University, Cleveland, OH, USA
| | - Mahek Shah
- Department of Cardiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sherry-Ann Brown
- Cardio-Oncology Program, Department of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brijesh Patel
- Heart and Vascular Institute, West Virginia University, 1 Medical Center Dr., Morgantown, WV, 26505, USA.
| |
Collapse
|
13
|
Gong K, Miao S, Yang L, Wu Y, Guo J, Chen W, Dai J, Du J, Xi S. Aaptamine attenuates the proliferation and progression of non-small cell lung carcinoma. PHARMACEUTICAL BIOLOGY 2020; 58:1044-1054. [PMID: 33027592 PMCID: PMC7580566 DOI: 10.1080/13880209.2020.1822420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
CONTEXT Aaptamine is a potent ocean-derived non-traditional drug candidate against human cancers. However, the underlying molecular mechanisms governing aaptamine-mediated repression of lung cancer cells remain largely undefined. OBJECTIVE To examine the inhibitory effect of aaptamine on proliferation and progression of non-small cell lung carcinoma (NSCLC) and dissect the potential mechanisms involved in its anticancer functions. MATERIALS AND METHODS In vitro assays of cell proliferation, cell cycle analysis, clonal formation, apoptosis and migration were performed to examine the inhibitory effects of aaptamine (8, 16 and 32 μg/mL) on NSCLC cells. The expression levels of proteins were analysed using western blotting analysis when cells were treated with a single drug or a combination treatment for 48 h. RESULTS Aaptamine significantly inhibited A549 and H1299 cells proliferation with IC50 values of 13.91 and 10.47 μg/mL. At the concentrations of 16 and 32 μg/mL, aaptamine significantly reduced capacities in clonogenicity, enhanced cellular apoptosis and decreased the motile and invasive cellular phenotype. In addition, aaptamine arrested cell cycle at G1 phase via selectively abating cell cycle regulation drivers (CDK2/4 and Cyclin D1/E). Western blotting results showed that aaptamine attenuated the protein expression of MMP-7, MMP-9 and upregulated the expression of cleaved-PARP and cleaved-caspase 3. Moreover, aaptamine inhibited PI3K/AKT/GSK3β signalling cascades through specifically degrading the phosphorylated AKT and GSK3β. DISCUSSION AND CONCLUSIONS Aaptamine retarded the proliferation and invasion of NSCLC cells by selectively targeting the pathway PI3K/AKT/GSK3β suggesting it as a potential chemotherapeutic agent for repressing tumorigenesis and progression of NSCLC in humans.
Collapse
Affiliation(s)
- Kaikai Gong
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, PR China
| | - Shuang Miao
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, PR China
| | - Lijuan Yang
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, PR China
| | - Yan Wu
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, PR China
| | - Jiwei Guo
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, PR China
| | - Weiwei Chen
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, PR China
| | - Juanjuan Dai
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, PR China
| | - Jing Du
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, PR China
- Jing Du Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, PR China
| | - Sichuan Xi
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, PR China
- CONTACT Sichuan Xi
| |
Collapse
|
14
|
Kristeleit R, Evans J, Molife LR, Tunariu N, Shaw H, Slater S, Haris NRM, Brown NF, Forster MD, Diamantis N, Rulach R, Greystoke A, Asghar U, Rata M, Anderson S, Bachmann F, Hannah A, Kaindl T, Lane HA, Larger PJ, Schmitt-Hoffmann A, Engelhardt M, Tzankov A, Plummer R, Lopez J. Phase 1/2a trial of intravenous BAL101553, a novel controller of the spindle assembly checkpoint, in advanced solid tumours. Br J Cancer 2020; 123:1360-1369. [PMID: 32741975 PMCID: PMC7591872 DOI: 10.1038/s41416-020-1010-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 06/29/2020] [Accepted: 07/16/2020] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND BAL101553 (lisavanbulin), the lysine prodrug of BAL27862 (avanbulin), exhibits broad anti-proliferative activity in human cancer models refractory to clinically relevant microtubule-targeting agents. METHODS This two-part, open-label, phase 1/2a study aimed to determine the maximum tolerated dose (MTD) and dose-limiting toxicities (DLTs) of 2-h infusion of BAL101553 in adults with advanced or recurrent solid tumours. The MTD was determined using a modified accelerated titration design in phase I. Patients received BAL101553 at the MTD and at lower doses in the phase 2a expansion to characterise safety and efficacy and to determine the recommended phase 2 dose (RP2D). RESULTS Seventy-three patients received BAL101553 at doses of 15-80 mg/m2 (phase 1, n = 24; phase 2a, n = 49). The MTD was 60 mg/m2; DLTs observed at doses ≥60 mg/m2 were reversible Grade 2-3 gait disturbance with Grade 2 peripheral sensory neuropathy. In phase 2a, asymptomatic myocardial injury was observed at doses ≥45 mg/m2. The RP2D for 2-h intravenous infusion was 30 mg/m2. The overall disease control rate was 26.3% in the efficacy population. CONCLUSIONS The RP2D for 2-h infusion of BAL101553 was well tolerated. Dose-limiting neurological and myocardial side effects were consistent with the agent's vascular-disrupting properties. CLINICAL TRIAL REGISTRATION EudraCT: 2010-024237-23.
Collapse
Affiliation(s)
- Rebecca Kristeleit
- Department of Oncology, Guys and St Thomas' NHS Foundation Trust, London, UK.
- National Institute for Health Research, University College London Hospitals Clinical Research Facility, London, UK.
| | - Jeffry Evans
- University of Glasgow, Glasgow, UK
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - L Rhoda Molife
- Drug Development Unit, The Royal Marsden Hospital and the Institute of Cancer Research, Sutton, UK
| | - Nina Tunariu
- Drug Development Unit, The Royal Marsden Hospital and the Institute of Cancer Research, Sutton, UK
| | - Heather Shaw
- National Institute for Health Research, University College London Hospitals Clinical Research Facility, London, UK
| | - Sarah Slater
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Noor R Md Haris
- Sir Bobby Robson Cancer Trials Research Centre, Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, UK
| | - Nicholas F Brown
- National Institute for Health Research, University College London Hospitals Clinical Research Facility, London, UK
| | - Martin D Forster
- National Institute for Health Research, University College London Hospitals Clinical Research Facility, London, UK
| | - Nikolaos Diamantis
- Drug Development Unit, The Royal Marsden Hospital and the Institute of Cancer Research, Sutton, UK
| | - Robert Rulach
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Alastair Greystoke
- Sir Bobby Robson Cancer Trials Research Centre, Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, UK
- Newcastle University, Newcastle upon Tyne, UK
| | - Uzma Asghar
- National Institute for Health Research, University College London Hospitals Clinical Research Facility, London, UK
| | - Mihaela Rata
- Radiotherapy and Imaging Unit, Institute of Cancer Research and Royal Marsden Hospital, Sutton, UK
| | | | - Felix Bachmann
- Basilea Pharmaceutica International Ltd, Basel, Switzerland
| | - Alison Hannah
- Oncology Clinical Trial Consulting, Sebastopol, CA, USA
| | - Thomas Kaindl
- Basilea Pharmaceutica International Ltd, Basel, Switzerland
| | - Heidi A Lane
- Basilea Pharmaceutica International Ltd, Basel, Switzerland
| | | | | | | | | | - Ruth Plummer
- Sir Bobby Robson Cancer Trials Research Centre, Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne, UK
- Newcastle University, Newcastle upon Tyne, UK
| | - Juanita Lopez
- Drug Development Unit, The Royal Marsden Hospital and the Institute of Cancer Research, Sutton, UK
| |
Collapse
|
15
|
Vincent J, Preston M, Mouchet E, Laugier N, Corrigan A, Boulanger J, Brown DG, Clark R, Wigglesworth M, Carter AP, Bullock SL. A High-Throughput Cellular Screening Assay for Small-Molecule Inhibitors and Activators of Cytoplasmic Dynein-1-Based Cargo Transport. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2020; 25:985-999. [PMID: 32436764 PMCID: PMC7116108 DOI: 10.1177/2472555220920581] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cytoplasmic dynein-1 (hereafter dynein) is a six-subunit motor complex that transports a variety of cellular components and pathogens along microtubules. Dynein's cellular functions are only partially understood, and potent and specific small-molecule inhibitors and activators of this motor would be valuable for addressing this issue. It has also been hypothesized that an inhibitor of dynein-based transport could be used in antiviral or antimitotic therapy, whereas an activator could alleviate age-related neurodegenerative diseases by enhancing microtubule-based transport in axons. Here, we present the first high-throughput screening (HTS) assay capable of identifying both activators and inhibitors of dynein-based transport. This project is also the first collaborative screening report from the Medical Research Council and AstraZeneca agreement to form the UK Centre for Lead Discovery. A cellular imaging assay was used, involving chemically controlled recruitment of activated dynein complexes to peroxisomes. Such a system has the potential to identify molecules that affect multiple aspects of dynein biology in vivo. Following optimization of key parameters, the assay was developed in a 384-well format with semiautomated liquid handling and image acquisition. Testing of more than 500,000 compounds identified both inhibitors and activators of dynein-based transport in multiple chemical series. Additional analysis indicated that many of the identified compounds do not affect the integrity of the microtubule cytoskeleton and are therefore candidates to directly target the transport machinery.
Collapse
Affiliation(s)
- John Vincent
- HTS, Discovery Sciences, Bio Pharmaceuticals R&D, AstraZeneca, Macclesfield, Cheshire, UK
| | - Marian Preston
- HTS, Discovery Sciences, Bio Pharmaceuticals R&D, AstraZeneca, Macclesfield, Cheshire, UK
| | - Elizabeth Mouchet
- HTS, Discovery Sciences, Bio Pharmaceuticals R&D, AstraZeneca, Macclesfield, Cheshire, UK
| | - Nicolas Laugier
- Division of Cell Biology, MRC Laboratory of Molecular Biology, Cambridge, Cambridgeshire, UK
| | - Adam Corrigan
- Quantitative Biology, Discovery Sciences, Bio Pharmaceuticals R&D, AstraZeneca, Cambridge, Cambridgeshire, UK
| | - Jérôme Boulanger
- Division of Cell Biology, MRC Laboratory of Molecular Biology, Cambridge, Cambridgeshire, UK
| | - Dean G Brown
- Hit Discovery, Discovery Sciences, Bio Pharmaceuticals R&D, AstraZeneca, Boston, USA
| | - Roger Clark
- Discovery Biology, Discovery Sciences, Bio Pharmaceuticals R&D, AstraZeneca, Cambridge, Cambridgeshire, UK
| | - Mark Wigglesworth
- HTS, Discovery Sciences, Bio Pharmaceuticals R&D, AstraZeneca, Macclesfield, Cheshire, UK
| | - Andrew P Carter
- Division of Structural Studies, MRC Laboratory of Molecular Biology, Cambridge, Cambridgeshire, UK
| | - Simon L Bullock
- Division of Cell Biology, MRC Laboratory of Molecular Biology, Cambridge, Cambridgeshire, UK
| |
Collapse
|
16
|
Ma X, Zhou L, Zheng S. Transcriptome analysis revealed key prognostic genes and microRNAs in hepatocellular carcinoma. PeerJ 2020; 8:e8930. [PMID: 32296612 PMCID: PMC7150540 DOI: 10.7717/peerj.8930] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/17/2020] [Indexed: 12/16/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. However, the molecular mechanisms involved in HCC remain unclear and are in urgent need of elucidation. Therefore, we sought to identify biomarkers in the prognosis of HCC through an integrated bioinformatics analysis. Methods Messenger RNA (mRNA) expression profiles were obtained from the Gene Expression Omnibus database and The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) for the screening of common differentially expressed genes (DEGs). Function and pathway enrichment analysis, protein-protein interaction network construction and key gene identification were performed. The significance of key genes in HCC was validated by overall survival analysis and immunohistochemistry. Meanwhile, based on TCGA data, prognostic microRNAs (miRNAs) were decoded using univariable and multivariable Cox regression analysis, and their target genes were predicted by miRWalk. Results Eleven hub genes (upregulated ASPM, AURKA, CCNB2, CDC20, PRC1 and TOP2A and downregulated AOX1, CAT, CYP2E1, CYP3A4 and HP) with the most interactions were considered as potential biomarkers in HCC and confirmed by overall survival analysis. Moreover, AURKA, PRC1, TOP2A, AOX1, CYP2E1, and CYP3A4 were considered candidate liver-biopsy markers for high risk of developing HCC and poor prognosis in HCC. Upregulation of hsa-mir-1269b, hsa-mir-518d, hsa-mir-548aq, hsa-mir-548f-1, and hsa-mir-6728, and downregulation of hsa-mir-139 and hsa-mir-4800 were determined to be risk factors of poor prognosis, and most of these miRNAs have strong potential to help regulate the expression of key genes. Conclusions This study undertook the first large-scale integrated bioinformatics analysis of the data from Illumina BeadArray platforms and the TCGA database. With a comprehensive analysis of transcriptional alterations, including mRNAs and miRNAs, in HCC, our study presented candidate biomarkers for the surveillance and prognosis of the disease, and also identified novel therapeutic targets at the molecular and pathway levels.
Collapse
Affiliation(s)
- Xi Ma
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China.,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China.,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, Zhejiang, China.,Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, Zhejiang, China.,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Sanchez-Carranza JN, González-Maya L, Razo-Hernández RS, Salas-Vidal E, Nolasco-Quintana NY, Clemente-Soto AF, García-Arizmendi L, Sánchez-Ramos M, Marquina S, Alvarez L. Achillin Increases Chemosensitivity to Paclitaxel, Overcoming Resistance and Enhancing Apoptosis in Human Hepatocellular Carcinoma Cell Line Resistant to Paclitaxel (Hep3B/PTX). Pharmaceutics 2019; 11:pharmaceutics11100512. [PMID: 31590262 PMCID: PMC6835644 DOI: 10.3390/pharmaceutics11100512] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/28/2019] [Accepted: 09/28/2019] [Indexed: 12/16/2022] Open
Abstract
Multidrug resistance (MDR) has become a major obstacle in the treatment of cancer, and is associated with mechanisms such as increased drug outflow, reduction of apoptosis, and/or altered drug metabolism. These problems can be mitigated by the coadministration of agents known as chemosensitizers, as they can reverse resistance to anticancer drugs and eventually resensitize cancer cells. We explore the chemosensitizing effect of Achillin, a guaianolide-type sesquiterpene lactone isolated from the Mexican medicinal plant Artemisia ludovisiana, to reverse MDR in Hep3B/PTX cells of hepatocellular carcinoma, which present resistance to paclitaxel (PTX). Achillin showed an important effect as chemosensitizer; indeed, the cytotoxic effect of PTX (25 nM) was enhanced, and the induction of G2/M phase cell cycle arrest and apoptosis were potentiated when combining with Achillin (100 μM). In addition, we observed that Achillin decreases P-gp levels and increases the intracellular retention of doxorubicin in Hep3B/PTX cells; in addition, homology structural modeling and molecular docking calculations predicted that Achillin interacts in two regions (M-site and R-site) of transporter drug efflux P-glycoprotein (P-gp). Our results suggest that the chemosensitizer effect demonstrated for Achillin could be associated with P-gp modulation. This work also provides useful information for the development of new therapeutic agents from guaianolide-type sesquiterpene lactones like Achillin.
Collapse
Affiliation(s)
- Jessica Nayelli Sanchez-Carranza
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| | - Leticia González-Maya
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| | - Rodrigo Said Razo-Hernández
- Centro de Investigación en Dinámica Celular-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Chamilpa, Cuernavaca 62209, Morelos, Mexico.
| | - Enrique Salas-Vidal
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca C.P. 62209, Morelos, Mexico.
| | - Ninfa Yaret Nolasco-Quintana
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
- Centro de Investigación en Dinámica Celular-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Chamilpa, Cuernavaca 62209, Morelos, Mexico.
| | - Aldo F Clemente-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa. Av. de las Américas y Blvd. Universitarios S/N, Culiacán 80010, Sinaloa, Mexico.
| | - Lucero García-Arizmendi
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| | - Mariana Sánchez-Ramos
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| | - Silvia Marquina
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| | - Laura Alvarez
- Centro de Investigaciones Químicas-IICBA, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa C.P. 62209, Cuernavaca, Mexico.
| |
Collapse
|
18
|
Joerger M, Stathis A, Metaxas Y, Hess D, Mantiero M, Mark M, Volden M, Kaindl T, Engelhardt M, Larger P, Lane H, Hafner P, Levy N, Stuedeli S, Sessa C, von Moos R. A Phase 1 study of BAL101553, a novel tumor checkpoint controller targeting microtubules, administered as 48-h infusion in adult patients with advanced solid tumors. Invest New Drugs 2019; 38:1067-1076. [PMID: 31471863 PMCID: PMC7340672 DOI: 10.1007/s10637-019-00850-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/21/2019] [Indexed: 02/04/2023]
Abstract
Purpose BAL101553, the prodrug of the microtubule-destabilizer BAL27862, previously showed signs of antitumor activity when administered as a 2-h infusion, but its use was limited by vascular toxicity. We investigated an alternative dosing strategy aimed at improving the safety profile of BAL101553. Methods This multicenter, open-label, Phase 1 dose-escalation study used a 3 + 3 design to determine the maximum tolerated dose (MTD), dose-limiting toxicities (DLTs), pharmacokinetics, and antitumor activity of BAL101553 administered as a 48-h IV infusion on Days 1, 8, and 15 of a 28-day cycle. Patients received oral BAL101553 on Days 15–21 of cycle 2 to assess oral bioavailability. Results BAL101553 was well tolerated at doses up to ≤70 mg/m2. Three grade 3 DLTs occurred: hypotension (70 mg/m2), hyponatremia and neutropenia (both 90 mg/m2). The MTD for 48-h IV BAL101553 was 70 mg/m2. At this dose level, the AUC for BAL27862 was 8580 ng.h/mL and the Cmax was 144 ng/mL. No apparent dose-related effects on blood pressure were observed with 48-h BAL101553 IV infusion. BAL27862 oral bioavailability was >80%. Conclusions Continuous 48-h IV BAL101553 infusion achieved higher exposure of the BAL27862 active metabolite than a 2-h infusion at the RP2D and did not cause vascular toxicity. Clinicaltrials.gov registration: NCT02895360.
Collapse
Affiliation(s)
| | | | - Yannis Metaxas
- Department of Medical Oncology, Cantonal Hospital Graubünden, Chur, Switzerland
| | - Dagmar Hess
- Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Mara Mantiero
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Michael Mark
- Department of Medical Oncology, Cantonal Hospital Graubünden, Chur, Switzerland
| | | | - Thomas Kaindl
- Basilea Pharmaceutica International Ltd, Grenzacherstrasse 487, PO Box, CH-4005, Basel, Switzerland.
| | - Marc Engelhardt
- Basilea Pharmaceutica International Ltd, Grenzacherstrasse 487, PO Box, CH-4005, Basel, Switzerland
| | - Patrice Larger
- Basilea Pharmaceutica International Ltd, Grenzacherstrasse 487, PO Box, CH-4005, Basel, Switzerland
| | - Heidi Lane
- Basilea Pharmaceutica International Ltd, Grenzacherstrasse 487, PO Box, CH-4005, Basel, Switzerland
| | - Peter Hafner
- Basilea Pharmaceutica International Ltd, Grenzacherstrasse 487, PO Box, CH-4005, Basel, Switzerland
| | - Nicole Levy
- Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | | | - Cristiana Sessa
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Roger von Moos
- Department of Medical Oncology, Cantonal Hospital Graubünden, Chur, Switzerland
| |
Collapse
|
19
|
Gargini R, Segura-Collar B, Sánchez-Gómez P. Novel Functions of the Neurodegenerative-Related Gene Tau in Cancer. Front Aging Neurosci 2019; 11:231. [PMID: 31551755 PMCID: PMC6736573 DOI: 10.3389/fnagi.2019.00231] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/13/2019] [Indexed: 12/14/2022] Open
Abstract
The analysis of global and comparative genomics between different diseases allows us to understand the key biological processes that explain the etiology of these pathologies. We have used this type of approach to evaluate the expression of several neurodegeneration-related genes on the development of tumors, particularly brain tumors of glial origin (gliomas), which are an aggressive and incurable type of cancer. We have observed that genes involved in Amyotrophic lateral sclerosis (ALS), as well as in Alzheimer’s and Parkinson’s diseases, correlate with better prognosis of gliomas. Within these genes, high Tau/MAPT expression shows the strongest correlation with several indicators of prolonged survival on glioma patients. Tau protein regulates microtubule stability and dynamics in neurons, although there have been reports of its expression in glial cells and also in gliomas. However, little is known about the regulation of Tau/MAPT transcription in tumors. Moreover, our in silico analysis indicates that this gene is also expressed in a variety of tumors, showing a general correlation with survival, although its function in cancer has not yet been addressed. Another remarkable aspect of Tau is its involvement in resistance to taxanes in various tumors types such as breast, ovarian and gastric carcinomas. This is due to the fact that taxanes have the same tubulin-binding site as Tau. In the present work we review the main knowledge about Tau function and expression in tumors, with a special focus on brain cancer. We will also speculate with the therapeutic implications of these findings.
Collapse
|
20
|
Wang Q, Arnst KE, Wang Y, Kumar G, Ma D, White SW, Miller DD, Li W, Li W. Structure-Guided Design, Synthesis, and Biological Evaluation of (2-(1 H-Indol-3-yl)-1 H-imidazol-4-yl)(3,4,5-trimethoxyphenyl) Methanone (ABI-231) Analogues Targeting the Colchicine Binding Site in Tubulin. J Med Chem 2019; 62:6734-6750. [PMID: 31251599 DOI: 10.1021/acs.jmedchem.9b00706] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
ABI-231 is a potent, orally bioavailable tubulin inhibitor that interacts with the colchicine binding site and is currently undergoing clinical trials for prostate cancer. Guided by the crystal structure of ABI-231 in complex with tubulin, we performed structure-activity relationship studies around the 3-indole moiety that led to the discovery of several potent ABI-231 analogues, most notably 10ab and 10bb. The crystal structures of 10ab and 10bb in complex with tubulin confirmed their improved molecular interactions to the colchicine site. In vitro, biological studies showed that new ABI-231 analogues disrupt tubulin polymerization, promote microtubule fragmentation, and inhibit cancer cell migration. In vivo, analogue 10bb not only significantly inhibits primary tumor growth and decreases tumor metastasis in melanoma xenograft models but also shows a significant ability to overcome paclitaxel resistance in a taxane-resistant PC-3/TxR model. In addition, pharmacological screening suggested that 10bb has a low risk of potential off-target function.
Collapse
Affiliation(s)
- Qinghui Wang
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Tennessee Health Science Center , Memphis , Tennessee 38163 , United States
| | - Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Tennessee Health Science Center , Memphis , Tennessee 38163 , United States
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| | - Gyanendra Kumar
- Department of Structural Biology , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 , United States
| | - Dejian Ma
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Tennessee Health Science Center , Memphis , Tennessee 38163 , United States
| | - Stephen W White
- Department of Structural Biology , St. Jude Children's Research Hospital , Memphis , Tennessee 38105 , United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Tennessee Health Science Center , Memphis , Tennessee 38163 , United States
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, West China Hospital , Sichuan University , Chengdu , Sichuan 610041 , China
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Tennessee Health Science Center , Memphis , Tennessee 38163 , United States
| |
Collapse
|
21
|
Arnst KE, Wang Y, Lei ZN, Hwang DJ, Kumar G, Ma D, Parke DN, Chen Q, Yang J, White SW, Seagroves TN, Chen ZS, Miller DD, Li W. Colchicine Binding Site Agent DJ95 Overcomes Drug Resistance and Exhibits Antitumor Efficacy. Mol Pharmacol 2019; 96:73-89. [PMID: 31043459 PMCID: PMC6553560 DOI: 10.1124/mol.118.114801] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 04/21/2019] [Indexed: 02/05/2023] Open
Abstract
Interfering with microtubule dynamics is a well-established strategy in cancer treatment; however, many microtubule-targeting agents are associated with drug resistance and adverse effects. Substantial evidence points to ATP-binding cassette (ABC) transporters as critical players in the development of resistance. Herein, we demonstrate the efficacy of DJ95 (2-(1H-indol-6-yl)-4-(3,4,5-trimethoxyphenyl)-1H-imidazo[4,5-c]pyridine), a novel tubulin inhibitor, in a variety of cancer cell lines, including malignant melanomas, drug-selected resistant cell lines, specific ABC transporter-overexpressing cell lines, and the National Cancer Institute 60 cell line panel. DJ95 treatment inhibited cancer cell migration, caused morphologic changes to the microtubule network foundation, and severely disrupted mitotic spindle formation of mitotic cells. The high-resolution crystal structure of DJ95 in complex with tubulin protein and the detailed molecular interactions confirmed its direct binding to the colchicine site. In vitro pharmacological screening of DJ95 using SafetyScreen44 (Eurofins Cerep-Panlabs) revealed no significant off-target interactions, and pharmacokinetic analysis showed that DJ95 was maintained at therapeutically relevant plasma concentrations for up to 24 hours in mice. In an A375 xenograft model in nude mice, DJ95 inhibited tumor growth and disrupted tumor vasculature in xenograft tumors. These results demonstrate that DJ95 is potent against a variety of cell lines, demonstrated greater potency to ABC transporter-overexpressing cell lines than existing tubulin inhibitors, directly targets the colchicine binding domain, exhibits significant antitumor efficacy, and demonstrates vascular-disrupting properties. Collectively, these data suggest that DJ95 has great potential as a cancer therapeutic, particularly for multidrug resistance phenotypes, and warrants further development. SIGNIFICANCE STATEMENT: Paclitaxel is a widely used tubulin inhibitor for cancer therapy, but its clinical efficacy is often limited by the development of multidrug resistance. In this study, we reported the preclinical characterization of a new tubulin inhibitor DJ95, and demonstrated its abilities to overcome paclitaxel resistance, disrupt tumor vasculature, and exhibit significant antitumor efficacy.
Collapse
Affiliation(s)
- Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Yuxi Wang
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Gyanendra Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Dejian Ma
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Deanna N Parke
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Qiang Chen
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Jinliang Yang
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Stephen W White
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Tiffany N Seagroves
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy (K.E.A., D.-J.H., D.M., D.D.M., W.L.), and Department of Pathology (D.N.P., T.N.S.), the University of Tennessee Health Science Center, Memphis, Tennessee; State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy (Y.W., Q.C., J.Y.), and Department of Respiratory Medicine (Y.W.), West China Hospital, Sichuan University, Chengdu, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York (Z.-N.L., Z.-S.C.); and Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee (G.K., S.W.W.)
| |
Collapse
|
22
|
Stark M, Assaraf YG. Structural recognition of tubulysin B derivatives by multidrug resistance efflux transporters in human cancer cells. Oncotarget 2018. [PMID: 28637003 PMCID: PMC5564821 DOI: 10.18632/oncotarget.18385] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Multidrug resistance (MDR) is a major hindrance to curative chemotherapy of various human malignancies. Hence, novel chemotherapeutics must be evaluated for their recognition by MDR efflux transporters. Herein we explored the cytotoxic activity of synthetic tubulysin B (Tub-B, EC1009) derivatives (Tub-B-hydrazide/EC0347 and Tub-B bis-ether/EC1820), and their recognition by the MDR efflux transporters P-glycoprotein 1 (P-gp), multidrug resistance-associated protein 1 (MRP1) and breast cancer resistance protein (BCRP). Originally isolated from Myxobacteria, tubulysins exhibited potent cytotoxic activity via microtubule depolymerization, and evaded recognition by these MDR efflux pumps. We show that subtle modifications in the natural Tub-B structure enhance its cytotoxicity and drug efflux efficiency. Whereas increasing the lipophilicity of Tub-B drugs enhanced their diffusion into the cell and consequently decreased the IC50 values (≥ 0.27 nM), increasing drug polarity enhanced their recognition by P-gp (>200-fold resistance in P-gp-overexpressing cells). Furthermore, restricting drug exposure time to the clinically relevant 4 h pulse, markedly enhanced efflux by P-gp, resulting in a 1000-fold increased resistance, which was further enhanced upon increased P-gp levels (i.e. an additional 3-fold increase in P-gp levels resulted in >6,000-fold resistance). The unique ability of EC1009 to evade recognition by MDR efflux pumps warrants drug development of tubulysin B derivatives as potent antitumor agents which overcome MDR in cancer.
Collapse
Affiliation(s)
- Michal Stark
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
23
|
Gutiérrez-Escobar AJ, Méndez-Callejas G. Interactome Analysis of Microtubule-targeting Agents Reveals Cytotoxicity Bases in Normal Cells. GENOMICS PROTEOMICS & BIOINFORMATICS 2017; 15:352-360. [PMID: 29246518 PMCID: PMC5828656 DOI: 10.1016/j.gpb.2017.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/17/2017] [Accepted: 04/13/2017] [Indexed: 12/29/2022]
Abstract
Cancer causes millions of deaths annually and microtubule-targeting agents (MTAs) are the most commonly-used anti-cancer drugs. However, the high toxicity of MTAs on normal cells raises great concern. Due to the non-selectivity of MTA targets, we analyzed the interaction network in a non-cancerous human cell. Subnetworks of fourteen MTAs were reconstructed and the merged network was compared against a randomized network to evaluate the functional richness. We found that 71.4% of the MTA interactome nodes are shared, which affects cellular processes such as apoptosis, cell differentiation, cell cycle control, stress response, and regulation of energy metabolism. Additionally, possible secondary targets were identified as client proteins of interphase microtubules. MTAs affect apoptosis signaling pathways by interacting with client proteins of interphase microtubules, suggesting that their primary targets are non-tumor cells. The paclitaxel and doxorubicin networks share essential topological axes, suggesting synergistic effects. This may explain the exacerbated toxicity observed when paclitaxel and doxorubicin are used in combination for cancer treatment.
Collapse
Affiliation(s)
- Andrés Julián Gutiérrez-Escobar
- Grupo de Investigaciones Biomédicas y Genética Aplicada - GIBGA, Universidad de Ciencias Aplicadas y Ambientales U.D.C.A., Bogotá 111166, Colombia.
| | - Gina Méndez-Callejas
- Grupo de Investigaciones Biomédicas y Genética Aplicada - GIBGA, Universidad de Ciencias Aplicadas y Ambientales U.D.C.A., Bogotá 111166, Colombia
| |
Collapse
|
24
|
Olazarán FE, García-Pérez CA, Bandyopadhyay D, Balderas-Rentería I, Reyes-Figueroa AD, Henschke L, Rivera G. Theoretical and experimental study of polycyclic aromatic compounds as β-tubulin inhibitors. J Mol Model 2017; 23:85. [PMID: 28214932 DOI: 10.1007/s00894-017-3256-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/23/2017] [Indexed: 12/14/2022]
Abstract
In this work, through a docking analysis of compounds from the ZINC chemical library on human β-tubulin using high performance computer cluster, we report new polycyclic aromatic compounds that bind with high energy on the colchicine binding site of β-tubulin, suggesting three new key amino acids. However, molecular dynamic analysis showed low stability in the interaction between ligand and receptor. Results were confirmed experimentally in in vitro and in vivo models that suggest that molecular dynamics simulation is the best option to find new potential β-tubulin inhibitors. Graphical abstract Bennett's acceptance ratio (BAR) method.
Collapse
Affiliation(s)
- Fabian E Olazarán
- Facultad de Ciencias Químicas. Av. Universidad s/n, Ciudad Universitaria, San Nicolás de los Garza, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, 64451, México
| | - Carlos A García-Pérez
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Boulevard del Maestro, s/n, Esq. Elías Piña, Reynosa, Tamualipas, Mexico, 88710
| | - Debasish Bandyopadhyay
- Department of Chemistry, The University of Texas Rio Grande Valley, 1201 West University Drive, Edinburg, TX, 78539, USA
| | - Isaias Balderas-Rentería
- Facultad de Ciencias Químicas. Av. Universidad s/n, Ciudad Universitaria, San Nicolás de los Garza, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, 64451, México
| | - Angel D Reyes-Figueroa
- Centro de Investigación y de Estudios Avanzados del Instituto Politecnico Nacional, Unidad Monterrey, Apodaca, Nuevo León, 66600, México
| | - Lars Henschke
- Department of Biology, University of Konstanz, Universitätsstraβe 10, 78457, Konstanz, Germany
| | - Gildardo Rivera
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Boulevard del Maestro, s/n, Esq. Elías Piña, Reynosa, Tamualipas, Mexico, 88710.
| |
Collapse
|
25
|
Zhang X, Zhao J, Gao X, Pei D, Gao C. Anthelmintic drug albendazole arrests human gastric cancer cells at the mitotic phase and induces apoptosis. Exp Ther Med 2017; 13:595-603. [PMID: 28352336 PMCID: PMC5348670 DOI: 10.3892/etm.2016.3992] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 11/10/2016] [Indexed: 12/13/2022] Open
Abstract
As microtubules have a vital function in the cell cycle, oncologists have developed microtubule inhibitors capable of preventing uncontrolled cell division, as in the case of cancer. The anthelmintic drug albendazole (ABZ) has been demonstrated to inhibit hepatocellular, ovarian and prostate cancer cells via microtubule targeting. However, its activity against human gastric cancer (GC) cells has remained to be determined. In the present study, ABZ was used to treat GC cells (MKN-45, SGC-7901 and MKN-28). A a CCK-8 cell proliferation assay was performed to assess the effects of ABZ on cell viability and cell cycle changes were assessed using flow cytometry. SGC-7901 cells were selected for further study, and flow cytometry was employed to determine the apoptotic rate, immunofluorescence analysis was employed to show changes of the microtubule structure as well as the subcellular localization and expression levels of cyclin B1, and western blot analysis was used to identify the dynamics of microtubule assembly. The expression levels of relevant proteins, including cyclin B1 and Cdc2, the two subunits of mitosis-promoting factor as well as apoptosis-asociated proteins were also assessed by western blot analysis. The results showed that ABZ exerted its anti-cancer activity in GC cell lines by disrupting microtubule formation and function to cause mitotic arrest, which is also associated with the accumulation of cyclin B1, and consequently induces apoptosis.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Oncology, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Jing Zhao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiangyang Gao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Dongsheng Pei
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Chao Gao
- Department of Oncology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
26
|
Jaroch K, Karolak M, Górski P, Jaroch A, Krajewski A, Ilnicka A, Sloderbach A, Stefański T, Sobiak S. Combretastatins: In vitro structure-activity relationship, mode of action and current clinical status. Pharmacol Rep 2016; 68:1266-1275. [PMID: 27686966 DOI: 10.1016/j.pharep.2016.08.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/18/2016] [Accepted: 08/23/2016] [Indexed: 02/08/2023]
Abstract
For the first time combretastatins were isolated from African willow tree Combretum Caffrum. Subsequent studies have shown the impact of combretastatin A4 phosphate, a water-soluble prodrug, on endothelial cells in tumor vascular system. The same effect was not observed in the vascular system. This selectivity is associated with combretastatins mechanism of action: binding to colchicine domain of microtubules, which affects the cytoskeleton functionality of immature endothelial cells. At the same time, combretastatins directly induce cell death via apoptosis and/or mitotic catastrophe pathways. The combination of both elements makes combretastatin an anticancer compound of high efficiency. The cis-configuration is crucial for its biological activity. To date, many derivatives were synthesized. The attempts to resolve spontaneous isomerization to less active trans-stilbene derivative are still in progress. This issue seems to be overcome by incorporation of the ethene bridge with heterocyclic moiety in combretastatins structure. This modification retains the cis-configuration and prevents isomerization. Nevertheless, combretastatin A4 phosphate disodium is still the most potent compound of this group. The combination therapy, which is the most effective treatment, includes combretastatin A4 phosphate (CA4P) and conventional chemotherapeutics and/or radiotherapy. CA4P is relatively well tolerated giving adverse events of moderate severity, which includes: nausea, vomiting, headache, and tumor pain. The aforementioned effects subside on the day of drug administration or on the following day.
Collapse
Affiliation(s)
- Karol Jaroch
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland.
| | - Maciej Karolak
- Department of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Przemysław Górski
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Alina Jaroch
- Department and Institute of Nutrition and Dietetics, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland; Department and Clinic of Geriatrics, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Adrian Krajewski
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | | | - Anna Sloderbach
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Tomasz Stefański
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Poznan University of Medical Sciences, Poznań, Poland; Department of Crystallography, Faculty of Chemistry, Adam Mickiewicz University, Poznań, Poland
| | - Stanisław Sobiak
- Department of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| |
Collapse
|
27
|
Lee HS, Lee NCO, Kouprina N, Kim JH, Kagansky A, Bates S, Trepel JB, Pommier Y, Sackett D, Larionov V. Effects of Anticancer Drugs on Chromosome Instability and New Clinical Implications for Tumor-Suppressing Therapies. Cancer Res 2016; 76:902-11. [PMID: 26837770 PMCID: PMC4827779 DOI: 10.1158/0008-5472.can-15-1617] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/09/2015] [Indexed: 12/21/2022]
Abstract
Whole chromosomal instability (CIN), manifested as unequal chromosome distribution during cell division, is a distinguishing feature of most cancer types. CIN is generally considered to drive tumorigenesis, but a threshold level exists whereby further increases in CIN frequency in fact hinder tumor growth. While this attribute is appealing for therapeutic exploitation, drugs that increase CIN beyond this therapeutic threshold are currently limited. In our previous work, we developed a quantitative assay for measuring CIN based on the use of a nonessential human artificial chromosome (HAC) carrying a constitutively expressed EGFP transgene. Here, we used this assay to rank 62 different anticancer drugs with respect to their effects on chromosome transmission fidelity. Drugs with various mechanisms of action, such as antimicrotubule activity, histone deacetylase inhibition, mitotic checkpoint inhibition, and targeting of DNA replication and damage responses, were included in the analysis. Ranking of the drugs based on their ability to induce HAC loss revealed that paclitaxel, gemcitabine, dactylolide, LMP400, talazoparib, olaparib, peloruside A, GW843682, VX-680, and cisplatin were the top 10 drugs demonstrating HAC loss at a high frequency. Therefore, identification of currently used compounds that greatly increase chromosome mis-segregation rates should expedite the development of new therapeutic strategies to target and leverage the CIN phenotype in cancer cells.
Collapse
Affiliation(s)
- Hee-Sheung Lee
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Nicholas C O Lee
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Natalay Kouprina
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jung-Hyun Kim
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Alex Kagansky
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Susan Bates
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jane B Trepel
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Yves Pommier
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Dan Sackett
- Program in Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland.
| | - Vladimir Larionov
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
28
|
Stabilizing versus destabilizing the microtubules: a double-edge sword for an effective cancer treatment option? Anal Cell Pathol (Amst) 2015; 2015:690916. [PMID: 26484003 PMCID: PMC4592889 DOI: 10.1155/2015/690916] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/29/2015] [Accepted: 09/01/2015] [Indexed: 11/17/2022] Open
Abstract
Microtubules are dynamic and structural cellular components involved in several cell functions, including cell shape, motility, and intracellular trafficking. In proliferating cells, they are essential components in the division process through the formation of the mitotic spindle. As a result of these functions, tubulin and microtubules are targets for anticancer agents. Microtubule-targeting agents can be divided into two groups: microtubule-stabilizing, and microtubule-destabilizing agents. The former bind to the tubulin polymer and stabilize microtubules, while the latter bind to the tubulin dimers and destabilize microtubules. Alteration of tubulin-microtubule equilibrium determines the disruption of the mitotic spindle, halting the cell cycle at the metaphase-anaphase transition and, eventually, resulting in cell death. Clinical application of earlier microtubule inhibitors, however, unfortunately showed several limits, such as neurological and bone marrow toxicity and the emergence of drug-resistant tumor cells. Here we review several natural and synthetic microtubule-targeting agents, which showed antitumor activity and increased efficacy in comparison to traditional drugs in various preclinical and clinical studies. Cryptophycins, combretastatins, ombrabulin, soblidotin, D-24851, epothilones and discodermolide were used in clinical trials. Some of them showed antiangiogenic and antivascular activity and others showed the ability to overcome multidrug resistance, supporting their possible use in chemotherapy.
Collapse
|
29
|
Gozzi GJ, Pires ADRA, Valdameri G, Rocha MEM, Martinez GR, Noleto GR, Acco A, Alves de Souza CE, Echevarria A, Moretto dos Reis C, Di Pietro A, Suter Correia Cadena SM. Selective Cytotoxicity of 1,3,4-Thiadiazolium Mesoionic Derivatives on Hepatocarcinoma Cells (HepG2). PLoS One 2015; 10:e0130046. [PMID: 26083249 PMCID: PMC4470815 DOI: 10.1371/journal.pone.0130046] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/15/2015] [Indexed: 12/26/2022] Open
Abstract
In this work, we evaluated the cytotoxicity of mesoionic 4-phenyl-5-(2-Y, 4-X or 4-X-cinnamoyl)-1,3,4-thiadiazolium-2-phenylamine chloride derivatives (MI-J: X=OH, Y=H; MI-D: X=NO2, Y=H; MI-4F: X=F, Y=H; MI-2,4diF: X=Y=F) on human hepatocellular carcinoma (HepG2), and non-tumor cells (rat hepatocytes) for comparison. MI-J, M-4F and MI-2,4diF reduced HepG2 viability by ~ 50% at 25 μM after 24-h treatment, whereas MI-D required a 50 μM concentration, as shown by 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. The cytotoxicity was confirmed with lactate dehydrogenase assay, of which activity was increased by 55, 24 and 16% for MI-J, MI-4F and MI-2,4diF respectively (at 25 μM after 24 h). To identify the death pathway related to cytotoxicity, the HepG2 cells treated by mesoionic compounds were labeled with both annexin V and PI, and analyzed by flow cytometry. All compounds increased the number of doubly-stained cells at 25 μM after 24 h: by 76% for MI-J, 25% for MI-4F and MI-2,4diF, and 11% for MI-D. It was also verified that increased DNA fragmentation occurred upon MI-J, MI-4F and MI-2,4diF treatments (by 12%, 9% and 8%, respectively, at 25 μM after 24 h). These compounds were only weakly, or not at all, transported by the main multidrug transporters, P-glycoprotein, ABCG2 and MRP1, and were able to slightly inhibit their drug-transport activity. It may be concluded that 1,3,4-thiadiazolium compounds, especially the hydroxy derivative MI-J, constitute promising candidates for future investigations on in-vivo treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Gustavo Jabor Gozzi
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | | | - Glaucio Valdameri
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Maria Eliane Merlin Rocha
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | - Glaucia Regina Martinez
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | | | - Alexandra Acco
- Departamento de Farmacologia, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | | | - Aurea Echevarria
- Departamento de Química, Universidade Federal Rural do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Attilio Di Pietro
- Equipe Labellisée Ligue 2014, BMSSI UMR 5086 CNRS/Université Lyon 1, IBCP, Lyon, France
| | | |
Collapse
|
30
|
Zulkipli IN, David SR, Rajabalaya R, Idris A. Medicinal Plants: A Potential Source of Compounds for Targeting Cell Division. Drug Target Insights 2015; 9:9-19. [PMID: 26106261 PMCID: PMC4468949 DOI: 10.4137/dti.s24946] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/07/2015] [Accepted: 04/20/2015] [Indexed: 01/28/2023] Open
Abstract
Modern medicinal plant drug discovery has provided pharmacologically active compounds targeted against a multitude of conditions and diseases, such as infection, inflammation, and cancer. To date, natural products from medicinal plants remain a solid niche as a source from which cancer therapies can be derived. Among other properties, one favorable characteristic of an anticancer drug is its ability to block the uncontrollable process of cell division, as cancer cells are notorious for their abnormal cell division. There are numerous other documented works on the potential anticancer activity of drugs derived from medicinal plants, and their effects on cell division are an attractive and growing therapeutic target. Despite this, there remains a vast number of unidentified natural products that are potentially promising sources for medical applications. This mini review aims to revise the current knowledge of the effects of natural plant products on cell division.
Collapse
Affiliation(s)
- Ihsan N Zulkipli
- PAP Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Sheba R David
- PAP Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Rajan Rajabalaya
- PAP Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| | - Adi Idris
- PAP Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei Darussalam
| |
Collapse
|
31
|
Li L, Liu D, Qiu ZX, Zhao S, Zhang L, Li WM. The prognostic role of mTOR and p-mTOR for survival in non-small cell lung cancer: a systematic review and meta-analysis. PLoS One 2015; 10:e0116771. [PMID: 25680114 PMCID: PMC4332670 DOI: 10.1371/journal.pone.0116771] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 12/13/2014] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES The mammalian target of rapamycin (mTOR) and phosphorylated mTOR (p-mTOR) are potential prognostic markers and therapeutic targets for non-small cell lung cancer (NSCLC). However, the association between mTOR/p-mTOR expression and NSCLC patients' prognosis remains controversial. Thus, a meta-analysis of existing studies evaluating the prognostic role of mTOR/p-mTOR expression for NSCLC was conducted. MATERIALS AND METHODS A systemically literature search was performed via Pubmed, Embase, Medline as well as CNKI (China National Knowledge Infrastructure). Studies were included that reported the hazard ratio (HR) and 95%CI for the association between mTOR/p-mTOR expression and NSCLC patients' survival. Random-effects model was used to pool HRs. RESULTS Ten eligible studies were included in this meta-analysis, with 4 about m-TOR and 7 about p-mTOR. For mTOR, the pooled HR of overall survival (OS) was 1.00 (95%CI 0.5 to 1.99) by univariate analysis and 1.22 (95%CI 0.53 to 2.82) by multivariate analysis. For p-mTOR, the pooled HR was 1.39 (95%CI 0.97 to 1.98) by univariate analysis and 1.42 (95%CI 0.56 to 3.60) by multivariate analysis. CONCLUSION The results indicated that no statistically significant association was found between mTOR/p-mTOR expression and NSCLC patients' prognosis.
Collapse
Affiliation(s)
- Lei Li
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Dan Liu
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Zhi-Xin Qiu
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Shuang Zhao
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Li Zhang
- Lab of Pathology, Department of Pathology, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Wei-Min Li
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| |
Collapse
|