1
|
Jang J, Koh B, Lee K. Discovery of benzimidazole-2-amide BNZ-111 as new tubulin inhibitor. Bioorg Med Chem Lett 2024; 113:129953. [PMID: 39270806 DOI: 10.1016/j.bmcl.2024.129953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/20/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
Methyl benzimidazole-2-carbamate anthelmintics are a class of oral drugs to treat parasitic worm infections via microtubule disruption for non-systemic indications and currently in use. In order to use for anticancer treatment, the new benzimidazoles needs to improve solubility and pharmacokinetic parameters while maintaining its cellular potency as for systemic drug. Structure-activity-relationship on the benzimidazole is thoroughly examined and a novel benzimidazole-2 propionamide BNZ-111 is identified having good oral exposure and bioavailability in rat. Molecular docking study suggests BNZ-111 have a specific binding mode to the β subunit of curved tubulin. BNZ-111 is potent to cancer cells and possesses good drug-like properties as oral drug. Especially, BNZ-111 is not a P-gp substrate and it demonstrates its efficacy over Paclitaxel-resistance tumor in vivo.
Collapse
Affiliation(s)
- Jiyoon Jang
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea
| | - Byumseok Koh
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea; Medicinal Chemistry & Pharmacology, Korea National University of Science & Technology, Daejeon 34113, South Korea.
| | - Kwangho Lee
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea; Medicinal Chemistry & Pharmacology, Korea National University of Science & Technology, Daejeon 34113, South Korea.
| |
Collapse
|
2
|
Tseng YW, Yang TJ, Hsu YL, Liu JH, Tseng YC, Hsu TW, Lu Y, Pan SH, Cheng TJR, Fang JM. Dual-targeting compounds possessing enhanced anticancer activity via microtubule disruption and histone deacetylase inhibition. Eur J Med Chem 2024; 265:116042. [PMID: 38141287 DOI: 10.1016/j.ejmech.2023.116042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/29/2023] [Accepted: 12/09/2023] [Indexed: 12/25/2023]
Abstract
Dual-targeting anticancer agents 4-29 are designed by combining the structural features of purine-type microtubule-disrupting compounds and HDAC inhibitors. A library of the conjugate compounds connected by appropriate linkers was synthesized and found to possess HDACs inhibitory activity and render microtubule fragmentation by activating katanin, a microtubule-severing protein. Among various zinc-binding groups, hydroxamic acid shows the highest inhibitory activity of Class I HDACs, which was also reconfirmed by three-dimensional quantitative structure-activity relationship (3D-QSAR) pharmacophore prediction. The purine-hydroxamate conjugates exhibit enhanced cytotoxicity against MDA-MB231 breast cancer cells, H1975 lung cancer cells, and various clinical isolated non-small-cell lung cancer cells with different epidermal growth factor receptor (EGFR) status. Pyridyl substituents could be used to replace the C2 and N9 phenyl moieties in the purine-type scaffold, which can help to improve the solubility under physiological conditions, thus increasing cytotoxicity. In mice treated with the purine-hydroxamate conjugates, the tumor growth rate was significantly reduced without causing toxic effects. Our study demonstrates the potential of the dual-targeting purine-hydroxamate compounds for cancer monotherapy.
Collapse
Affiliation(s)
- Yu-Wei Tseng
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan
| | - Tsung-Jung Yang
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan
| | - Yuan-Ling Hsu
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Jyung-Hurng Liu
- Graduate Institute of Genomics and Bioinformatics, College of Life Sciences, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Yin-Chen Tseng
- The Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Tse-Wei Hsu
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan
| | - Yueh Lu
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan
| | - Szu-Hua Pan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, 100, Taiwan; Doctoral Degree Program of Translational Medicine, National Taiwan University, Taipei, 100, Taiwan; Genome and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei, 100, Taiwan.
| | | | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan; The Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan.
| |
Collapse
|
3
|
Huynh H, Ng WH, Soo KC. Everolimus Acts in Synergy with Vinorelbine to Suppress the Growth of Hepatocellular Carcinoma. Int J Mol Sci 2023; 25:17. [PMID: 38203186 PMCID: PMC10779360 DOI: 10.3390/ijms25010017] [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: 11/15/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a challenging cancer to treat, as traditional chemotherapies have shown limited effectiveness. The mammalian target of rapamycin/sirolimus (mTOR) and microtubules are prominent druggable targets for HCC. In this study, we demonstrated that co-targeting mTOR using mTOR inhibitors (everolimus and sirolimus) along with the microtubule inhibitor vinorelbine yielded results superior to those of the monotherapies in HCC PDX models. Our research showed that the vinorelbine arrests cells at the mitotic phase, induces apoptosis, and normalizes tumor blood vessels but upregulates survivin and activates the mTOR/p70S6K/4EBP1 pathway. The addition of the everolimus significantly improved the tumor response to the vinorelbine, leading to improved overall survival (OS) in most tested orthotopic HCC PDX models. The mechanistic investigation revealed that this marked antitumor effect was accompanied by the downregulations of mTOR targets (p-p70S6K, p-4EBP1, and p-S6K); several key cell-cycle regulators; and the antiapoptotic protein survivin. These effects did not compromise the normalization of the blood vessels observed in response to the vinorelbine in the vinorelbine-sensitive PDX models or to the everolimus in the everolimus-sensitive PDX models. The combination of the everolimus and vinorelbine (everolimus/vinorelbine) also promoted apoptosis with minimal toxicity. Given the cost-effectiveness and established effectiveness of everolimus, and especially sirolimus, this strategy warrants further investigation in early-phase clinical trials.
Collapse
Affiliation(s)
- Hung Huynh
- Laboratory of Molecular Endocrinology, National Cancer Centre Singapore, Singapore 168583, Singapore;
| | - Wai Har Ng
- Laboratory of Molecular Endocrinology, National Cancer Centre Singapore, Singapore 168583, Singapore;
| | - Khee Chee Soo
- Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, Singapore 168583, Singapore
| |
Collapse
|
4
|
Hu Y, Yang L, Lai Y. Recent findings regarding the synergistic effects of emodin and its analogs with other bioactive compounds: Insights into new mechanisms. Biomed Pharmacother 2023; 162:114585. [PMID: 36989724 DOI: 10.1016/j.biopha.2023.114585] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
CONTEXT Emodin is a natural bioactive ingredient mainly extracted from traditional Chinese herbs. Increasing lines of evidence suggest that emodin and its analogs exert notable synergistic pharmacological effects with other bioactive compounds. OBJECTIVE This review provides an overview of the pharmacological activity of emodin and its analogs in combination with other physiologically active substances, describes the related molecular mechanisms, and discusses future prospects in this field. METHODS Information from multiple scientific databases, such as PubMed, the China Knowledge Resource Integrated Database from the China National Knowledge Infrastructure (CNKI), the Web of Science, Google Scholar, and Baidu Scholar, was collected between January 2006 and August 2022. The subject terms used in the literature search were emodin, pharmaceutical activities, analogs, aloe emodin, rhein, and synergistic effects. RESULTS The comprehensive literature analysis suggested that combinations of emodin or its analogs with other bioactive compounds exert notable synergistic anticancer, anti-inflammatory, and antimicrobial effects and that such combinations improve glucose and lipid metabolism and central nervous system diseases. DISCUSSION AND CONCLUSIONS Further assessments of the dose-effect relationship and the differences in the efficacy of emodin or its analogs with other bioactive compounds among various modes of administration are needed, and a drug safety evaluation of these combinations needs to be carefully performed. Future studies should also focus on determining the optimal drug combinations for specific diseases.
Collapse
|
5
|
Jang J, Lee K, Koh B. Investigation of benzimidazole anthelmintics as oral anticancer agents. B KOREAN CHEM SOC 2022. [DOI: 10.1002/bkcs.12519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jiyoon Jang
- Therapeutics and Biotechnology Division Korea Research Institute of Chemical Technology Daejeon South Korea
- Department of Chemistry Sungkyunkwan University Suwon South Korea
| | - Kwangho Lee
- Therapeutics and Biotechnology Division Korea Research Institute of Chemical Technology Daejeon South Korea
- Medicinal Chemistry & Pharmacology University of Science & Technology Daejeon South Korea
| | - Byumseok Koh
- Therapeutics and Biotechnology Division Korea Research Institute of Chemical Technology Daejeon South Korea
| |
Collapse
|
6
|
Peerzada MN, Hamel E, Bai R, Supuran CT, Azam A. Deciphering the key heterocyclic scaffolds in targeting microtubules, kinases and carbonic anhydrases for cancer drug development. Pharmacol Ther 2021; 225:107860. [PMID: 33895188 DOI: 10.1016/j.pharmthera.2021.107860] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 12/17/2022]
Abstract
Heterocyclic scaffolds are widely utilized for drug design by taking into account the molecular structure of therapeutic targets that are related to a broad spectrum of ailments, including tumors. Such compounds display various covalent and non-covalent interactions with the specific residues of the target proteins while causing their inhibition. There is a substantial number of heterocyclic compounds approved for cancer treatment, and these compounds function by interacting with different therapeutic targets involved in tumorogenesis. In this review, we trace and emphasize the privileged heterocyclic pharmacophores that have immense potency against several essential chemotherapeutic tumor targets: microtubules, kinases and carbonic anhydrases. Potent compounds currently undergoing pre-clinical and clinical studies have also been assessed for ascertaining the effective class of chemical scaffolds that have significant therapeutic potential against multiple malignancies. In addition, we also describe briefly the role of heterocyclic compounds in various chemotherapy regimens. The optimized molecular hybridization of delineated motifs may result in the discovery of more active anticancer therapeutics and circumvent the development of resistance by specific targets in the future.
Collapse
Affiliation(s)
- Mudasir Nabi Peerzada
- Medicinal Chemistry Research Laboratory, Department of Chemistry, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019 Sesto Fiorentino, Florence, Italy.
| | - Amir Azam
- Medicinal Chemistry Research Laboratory, Department of Chemistry, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
7
|
A lncRNA landscape in breast cancer reveals a potential role for AC009283.1 in proliferation and apoptosis in HER2-enriched subtype. Sci Rep 2020; 10:13146. [PMID: 32753692 PMCID: PMC7403317 DOI: 10.1038/s41598-020-69905-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 07/19/2020] [Indexed: 12/23/2022] Open
Abstract
Breast cancer is the most commonly diagnosed neoplasm in women worldwide with a well-recognized heterogeneous pathology, classified into four molecular subtypes: Luminal A, Luminal B, HER2-enriched and Basal-like, each one with different biological and clinical characteristics. Long non-coding RNAs (lncRNAs) represent 33% of the human transcriptome and play critical roles in breast carcinogenesis, but most of their functions are still unknown. Therefore, cancer research could benefit from continued exploration into the biology of lncRNAs in this neoplasm. We characterized lncRNA expression portraits in 74 breast tumors belonging to the four molecular subtypes using transcriptome microarrays. To infer the biological role of the deregulated lncRNAs in the molecular subtypes, we performed co-expression analysis of lncRNA-mRNA and gene ontology analysis. We identified 307 deregulated lncRNAs in tumor compared to normal tissue and 354 deregulated lncRNAs among the different molecular subtypes. Through co-expression analysis between lncRNAs and protein-coding genes, along with gene enrichment analysis, we inferred the potential function of the most deregulated lncRNAs in each molecular subtype, and independently validated our results taking advantage of TCGA data. Overexpression of the AC009283.1 was observed in the HER2-enriched subtype and it is localized in an amplification zone at chromosome 17q12, suggesting it to be a potential tumorigenic lncRNA. The functional role of lncRNA AC009283.1 was examined through loss of function assays in vitro and determining its impact on global gene expression. These studies revealed that AC009283.1 regulates genes involved in proliferation, cell cycle and apoptosis in a HER2 cellular model. We further confirmed these findings through ssGSEA and CEMITool analysis in an independent HER2-amplified breast cancer cohort. Our findings suggest a wide range of biological functions for lncRNAs in each breast cancer molecular subtype and provide a basis for their biological and functional study, as was conducted for AC009283.1, showing it to be a potential regulator of proliferation and apoptosis in the HER2-enriched subtype.
Collapse
|
8
|
Zeng Z, Vo AH, Mao C, Clare SE, Khan SA, Luo Y. Cancer classification and pathway discovery using non-negative matrix factorization. J Biomed Inform 2019; 96:103247. [PMID: 31271844 PMCID: PMC6697569 DOI: 10.1016/j.jbi.2019.103247] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/23/2019] [Accepted: 07/01/2019] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Extracting genetic information from a full range of sequencing data is important for understanding disease. We propose a novel method to effectively explore the landscape of genetic mutations and aggregate them to predict cancer type. DESIGN We applied non-smooth non-negative matrix factorization (nsNMF) and support vector machine (SVM) to utilize the full range of sequencing data, aiming to better aggregate genetic mutations and improve their power to predict disease type. More specifically, we introduce a novel classifier to distinguish cancer types using somatic mutations obtained from whole-exome sequencing data. Mutations were identified from multiple cancers and scored using SIFT, PP2, and CADD, and collapsed at the individual gene level. nsNMF was then applied to reduce dimensionality and obtain coefficient and basis matrices. A feature matrix was derived from the obtained matrices to train a classifier for cancer type classification with the SVM model. RESULTS We have demonstrated that the classifier was able to distinguish four cancer types with reasonable accuracy. In five-fold cross-validations using mutation counts as features, the average prediction accuracy was 80% (SEM = 0.1%), significantly outperforming baselines and outperforming models using mutation scores as features. CONCLUSION Using the factor matrices derived from the nsNMF, we identified multiple genes and pathways that are significantly associated with each cancer type. This study presents a generic and complete pipeline to study the associations between somatic mutations and cancers. The proposed method can be adapted to other studies for disease status classification and pathway discovery.
Collapse
Affiliation(s)
- Zexian Zeng
- Department of Preventive Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Andy H Vo
- Committee on Developmental Biology and Regenerative Medicine, The University of Chicago, Chicago, IL, USA
| | - Chengsheng Mao
- Department of Preventive Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Susan E Clare
- Department of Surgery, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| | - Seema A Khan
- Department of Surgery, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| | - Yuan Luo
- Department of Preventive Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
9
|
Kuo TC, Li LW, Pan SH, Fang JM, Liu JH, Cheng TJ, Wang CJ, Hung PF, Chen HY, Hong TM, Hsu YL, Wong CH, Yang PC. Purine-Type Compounds Induce Microtubule Fragmentation and Lung Cancer Cell Death through Interaction with Katanin. J Med Chem 2016; 59:8521-34. [DOI: 10.1021/acs.jmedchem.6b00797] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Ting-Chun Kuo
- Ph.D.
Program in Translational Medicine, National Taiwan University and Academia Sinica, Taipei, Taiwan
| | - Ling-Wei Li
- Department
of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Szu-Hua Pan
- Ph.D.
Program in Translational Medicine, National Taiwan University and Academia Sinica, Taipei, Taiwan
- Graduate
Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Genome
and Systems Biology Degree Program, National Taiwan University and Academia Sinica, Taipei 106, Taiwan
| | - Jim-Min Fang
- Department
of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Jyung-Hurng Liu
- Department
of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
- Institute
of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
- Agricultural
Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
- Rong
Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Ting-Jen Cheng
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chia-Jen Wang
- Department
of Internal Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Institute
of Stem Cell and Translational Cancer Research, Chang Gung Memorial HospitalTaipei 105, Taiwan
| | - Pei-Fang Hung
- Department
of Internal Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Hsuan-Yu Chen
- Institute
of Statistical Science, Academia Sinica, Taipei 115, Taiwan
| | - Tse-Ming Hong
- Institute
of Clinical Medicine, National Cheng Kung University College of Medicine, Tainan 701, Taiwan
| | - Yuan-Ling Hsu
- Graduate
Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chi-Huey Wong
- Genomics
Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Pan-Chyr Yang
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- NTU
Center for Genomic Medicine, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|
10
|
Zynda ER, Matveev V, Makhanov M, Chenchik A, Kandel ES. Protein kinase A type II-α regulatory subunit regulates the response of prostate cancer cells to taxane treatment. Cell Cycle 2015; 13:3292-301. [PMID: 25485509 DOI: 10.4161/15384101.2014.949501] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In the last decade taxane-based therapy has emerged as a standard of care for hormone-refractory prostate cancer. Nevertheless, a significant fraction of tumors show no appreciable response to the treatment, while the others develop resistance and recur. Despite years of intense research, the mechanisms of taxane resistance in prostate cancer and other malignancies are poorly understood and remain a topic of intense investigation. We have used improved mutagenesis via random insertion of a strong promoter to search for events, which enable survival of prostate cancer cells after Taxol exposure. High-throughput mapping of the integration sites pointed to the PRKAR2A gene, which codes for a type II-α regulatory subunit of protein kinase A, as a candidate modulator of drug response. Both full-length and N-terminally truncated forms of the PRKAR2A gene product markedly increased survival of prostate cancer cells lines treated with Taxol and Taxotere. Suppression of protein kinase A enzymatic activity is the likely mechanism of action of the overexpressed proteins. Accordingly, protein kinase A inhibitor PKI (6-22) amide reduced toxicity of Taxol to prostate cancer cells. Our findings support the role of protein kinase A and its constituent proteins in cell response to chemotherapy.
Collapse
Affiliation(s)
- Evan R Zynda
- a Department of Cell Stress Biology ; Roswell Park Cancer Institute ; Buffalo , NY USA
| | | | | | | | | |
Collapse
|
11
|
Loong HH, Yeo W. Microtubule-targeting agents in oncology and therapeutic potential in hepatocellular carcinoma. Onco Targets Ther 2014; 7:575-585. [PMID: 24790457 PMCID: PMC3999274 DOI: 10.2147/ott.s46019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In mammalian cells, microtubules are present both in interphase and dividing cells. In the latter, microtubules forming the mitotic spindle are highly dynamic and exquisitely sensitive to therapeutic inhibitors. Developed to alter microtubule function, microtubule-binding agents have been proven to be highly active as an anticancer treatment. Significant development of microtubule-binding agents has taken place in recent years, with newer anti-tubulin agents now showing novel properties of enhanced tumor specificity, reduced neurotoxicity, and insensitivity to chemoresistance mechanisms. Hepatocellular carcinoma remains one of the most difficult cancers to treat, with chemotherapies being relatively ineffective. There is now evidence to suggest that microtubule-binding agents may be effective in the treatment of hepatocellular carcinoma, especially when used in combination with mammalian target of rapamycin inhibitors. Preclinical models have suggested that the latter may be able to overcome resistance to microtubule binding agents. In this review article, recent developments of novel microtubule binding agents and their relevance to the treatment of hepatocellular carcinoma will be discussed.
Collapse
Affiliation(s)
- Herbert H Loong
- Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Hong Kong Cancer Institute, State Key Laboratory in Oncology in South China, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Winnie Yeo
- Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Hong Kong Cancer Institute, State Key Laboratory in Oncology in South China, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| |
Collapse
|
12
|
Giovinazzi S, Morozov VM, Summers MK, Reinhold WC, Ishov AM. USP7 and Daxx regulate mitosis progression and taxane sensitivity by affecting stability of Aurora-A kinase. Cell Death Differ 2013; 20:721-31. [PMID: 23348568 DOI: 10.1038/cdd.2012.169] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
A large number of patients are resistant to taxane-based chemotherapy. Functional mitotic checkpoints are essential for taxane sensitivity. Thus, mitotic regulators are potential markers for therapy response and could be targeted for anticancer therapy. In this study, we identified a novel function of ubiquitin (Ub)-specific processing protease-7 (USP7) that interacts and cooperates with protein death domain-associated protein (Daxx) in the regulation of mitosis and taxane resistance. Depletion of USP7 impairs mitotic progression, stabilizes cyclin B and reduces stability of the mitotic E3 Ub ligase, checkpoint with forkhead and Ring-finger (CHFR). Consequently, cells with depleted USP7 accumulate Aurora-A kinase, a CHFR substrate, thus elevating multipolar mitoses. We further show that these effects are independent of the USP7 substrate p53. Thus, USP7 and Daxx are necessary to regulate proper execution of mitosis, partially via regulation of CHFR and Aurora-A kinase stability. Results from colony formation assay, in silico analysis across the NCI60 platform and in breast cancer patients suggest that USP7 levels inversely correlate with response to taxanes, pointing at the USP7 protein as a potential predictive factor for taxane response in cancer patients. In addition, we demonstrated that inhibition of Aurora-A attenuates USP7-mediated taxane resistance, suggesting that combinatorial drug regimens of Taxol and Aurora-A inhibitors may improve the outcome of chemotherapy response in cancer patients resistant to taxane treatment. Finally, our study offers novel insights on USP7 inhibition as cancer therapy.
Collapse
Affiliation(s)
- S Giovinazzi
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | |
Collapse
|
13
|
Sciani JM, de-Sá-Júnior PL, Ferreira AK, Pereira A, Antoniazzi MM, Jared C, Pimenta DC. Cytotoxic and antiproliferative effects of crude amphibian skin secretions on breast tumor cells. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.bionut.2012.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
14
|
Fitzgerald DP, Emerson DL, Qian Y, Anwar T, Liewehr DJ, Steinberg SM, Silberman S, Palmieri D, Steeg PS. TPI-287, a new taxane family member, reduces the brain metastatic colonization of breast cancer cells. Mol Cancer Ther 2012; 11:1959-67. [PMID: 22622283 PMCID: PMC7489285 DOI: 10.1158/1535-7163.mct-12-0061] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Brain metastases of breast and other cancers remain resistant to chemotherapeutic regimens that are effective systemically, in part due to the blood-brain barrier. We report that TPI-287, a new microtubule-stabilizing agent, displays in vitro cytotoxic activity similar to taxanes and epothilones. Unlike the taxanes, TPI-287 is permeable through the blood-brain barrier. Brain-to-plasma ratios of TPI-287 after a single injection typically exceeded one and were as high as 63.8 in the rat and 14.1 in the mouse. A brain-tropic derivative of the MDA-MB-231 triple-negative breast cancer cell line, 231-BR, was used to test whether TPI-287 may be efficacious at preventing or treating brain metastases. TPI-287 had growth inhibitory effects comparable with paclitaxel when 231-BR tumor cells were injected into the mammary fat pad. Brain metastatic colonization was determined by intracardiac injection of 231-BR cells, with treatment beginning on day 3 to 4 postinjection, culminating in a histologic count of brain metastases in brains necropsied days 25 to 28 postinjection. In this assay, paclitaxel, ixabepilone, and nab paclitaxel did not have significant inhibitory activity. TPI-287 was ineffective in the same assay using a 6 mg/kg every week schedule; however an 18 mg/kg dose delivered on days 3, 7, and 11 significantly reduced the outgrowth of brain metastases (55% reduction, P = 0.028) and reduced proliferation in brain metastases (16% reduction, P = 0.008). When TPI-287 treatment was delayed until days 18, 22, and 26 postinjection, efficacy was reduced (17% reduction, not significant). These data suggest that TPI-287 may have efficacy when administered early in the course of the disease.
Collapse
Affiliation(s)
- Daniel P. Fitzgerald
- Women's Cancers Section, Laboratory of Molecular Pharmacology, National Cancer Institute, Bethesda, Maryland
| | | | - Yongzhen Qian
- Laboratory Animal Sciences Program, Science Applications International Corporation, National Cancer Institute, Frederick, Maryland
| | - Talha Anwar
- Women's Cancers Section, Laboratory of Molecular Pharmacology, National Cancer Institute, Bethesda, Maryland
| | - David J. Liewehr
- Biostatistics and Data Management Section, National Cancer Institute, Bethesda, Maryland
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, National Cancer Institute, Bethesda, Maryland
| | | | - Diane Palmieri
- Women's Cancers Section, Laboratory of Molecular Pharmacology, National Cancer Institute, Bethesda, Maryland
| | - Patricia S. Steeg
- Women's Cancers Section, Laboratory of Molecular Pharmacology, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
15
|
Muñoz-Couselo E, Pérez-García J, Cortés J. Eribulin mesylate as a microtubule inhibitor for treatment of patients with metastatic breast cancer. Onco Targets Ther 2011; 4:185-92. [PMID: 22162924 PMCID: PMC3233277 DOI: 10.2147/ott.s16392] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Metastatic breast cancer (MBC) remains an incurable disease, with the goals of care aimed at maximizing the patient's duration and quality of life. Treatment options for MBC have become more efficacious and numerous. In addition to endocrine and chemotherapy agents, a number of targeted agents, including trastuzumab and bevacizumab, have further enhanced the landscape of therapeutic options. Eribulin mesylate (E7389) is a nontaxane microtubule dynamics inhibitor, and a structurally simplified synthetic analog of the natural marine product, halichondrin B, with a novel mechanism of action that has shown antitumor activity in pretreated MBC. Eribulin has shown a manageable tolerability profile in Phase I-II clinical trials and an improvement in overall survival compared with treatment of physician's choice, without relevant toxicities in a recently published Phase III trial. This review will focus on eribulin as a new active agent for MBC and its role in the management of breast disease.
Collapse
Affiliation(s)
- Eva Muñoz-Couselo
- Breast Cancer Unit, Vall d’Hebron Institute of Oncology, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José Pérez-García
- Breast Cancer Unit, Vall d’Hebron Institute of Oncology, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Javier Cortés
- Breast Cancer Unit, Vall d’Hebron Institute of Oncology, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|