1
|
Abdelrady YA, Thabet HS, Sayed AM. The future of metronomic chemotherapy: experimental and computational approaches of drug repurposing. Pharmacol Rep 2024:10.1007/s43440-024-00662-w. [PMID: 39432183 DOI: 10.1007/s43440-024-00662-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
Metronomic chemotherapy (MC), long-term continuous administration of anticancer drugs, is gaining attention as an alternative to the traditional maximum tolerated dose (MTD) chemotherapy. By combining MC with other treatments, the therapeutic efficacy is enhanced while minimizing toxicity. MC employs multiple mechanisms, making it a versatile approach against various cancers. However, drug resistance limits the long-term effectiveness of MC, necessitating ongoing development of anticancer drugs. Traditional drug discovery is lengthy and costly due to processes like target protein identification, virtual screening, lead optimization, and safety and efficacy evaluations. Drug repurposing (DR), which screens FDA-approved drugs for new uses, is emerging as a cost-effective alternative. Both experimental and computational methods, such as protein binding assays, in vitro cytotoxicity tests, structure-based screening, and several types of association analyses (Similarity-Based, Network-Based, and Target Gene), along with retrospective clinical analyses, are employed for virtual screening. This review covers the mechanisms of MC, its application in various cancers, DR strategies, examples of repurposed drugs, and the associated challenges and future directions.
Collapse
Affiliation(s)
- Yousef A Abdelrady
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104, Freiburg, Germany
| | - Hayam S Thabet
- Microbiology Department, Faculty of Veterinary Medicine, Assiut University, Asyut, 71526, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Asyut, 71516, Egypt
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Kingdom of Saudi Arabia
| |
Collapse
|
2
|
Stauffer PE, Brinkley J, Jacobson DA, Quaranta V, Tyson DR. Purinergic Ca 2+ Signaling as a Novel Mechanism of Drug Tolerance in BRAF-Mutant Melanoma. Cancers (Basel) 2024; 16:2426. [PMID: 39001489 PMCID: PMC11240618 DOI: 10.3390/cancers16132426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Drug tolerance is a major cause of relapse after cancer treatment. Despite intensive efforts, its molecular basis remains poorly understood, hampering actionable intervention. We report a previously unrecognized signaling mechanism supporting drug tolerance in BRAF-mutant melanoma treated with BRAF inhibitors that could be of general relevance to other cancers. Its key features are cell-intrinsic intracellular Ca2+ signaling initiated by P2X7 receptors (purinergic ligand-gated cation channels) and an enhanced ability for these Ca2+ signals to reactivate ERK1/2 in the drug-tolerant state. Extracellular ATP, virtually ubiquitous in living systems, is the ligand that can initiate Ca2+ spikes via P2X7 channels. ATP is abundant in the tumor microenvironment and is released by dying cells, ironically implicating treatment-initiated cancer cell death as a source of trophic stimuli that leads to ERK reactivation and drug tolerance. Such a mechanism immediately offers an explanation of the inevitable relapse after BRAFi treatment in BRAF-mutant melanoma and points to actionable strategies to overcome it.
Collapse
Affiliation(s)
- Philip E. Stauffer
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jordon Brinkley
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - David A. Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
| | - Vito Quaranta
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Darren R. Tyson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
3
|
Stauffer PE, Brinkley J, Jacobson D, Quaranta V, Tyson DR. Purinergic Ca 2+ signaling as a novel mechanism of drug tolerance in BRAF mutant melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.03.565532. [PMID: 37961267 PMCID: PMC10635130 DOI: 10.1101/2023.11.03.565532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Drug tolerance is a major cause of relapse after cancer treatment. In spite of intensive efforts1-9, its molecular basis remains poorly understood, hampering actionable intervention. We report a previously unrecognized signaling mechanism supporting drug tolerance in BRAF-mutant melanoma treated with BRAF inhibitors that could be of general relevance to other cancers. Its key features are cell-intrinsic intracellular Ca2+ signaling initiated by P2X7 receptors (purinergic ligand-gated cation channels), and an enhanced ability for these Ca2+ signals to reactivate ERK1/2 in the drug-tolerant state. Extracellular ATP, virtually ubiquitous in living systems, is the ligand that can initiate Ca2+ spikes via P2X7 channels. ATP is abundant in the tumor microenvironment and is released by dying cells, ironically implicating treatment-initiated cancer cell death as a source of trophic stimuli that leads to ERK reactivation and drug tolerance. Such a mechanism immediately offers an explanation of the inevitable relapse after BRAFi treatment in BRAF-mutant melanoma, and points to actionable strategies to overcome it.
Collapse
Affiliation(s)
- Philip E Stauffer
- Department of Pharmacology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Jordon Brinkley
- Department of Pharmacology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - David Jacobson
- Departments of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Vito Quaranta
- Department of Pharmacology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
- Department of Biochemistry, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Darren R Tyson
- Department of Pharmacology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| |
Collapse
|
4
|
Kataria A, Tyagi S. Domain architecture and protein-protein interactions regulate KDM5A recruitment to the chromatin. Epigenetics 2023; 18:2268813. [PMID: 37838974 PMCID: PMC10578193 DOI: 10.1080/15592294.2023.2268813] [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: 05/18/2023] [Accepted: 10/01/2023] [Indexed: 10/17/2023] Open
Abstract
Tri-methylation of Histone 3 lysine 4 (H3K4) is an important epigenetic modification whose deposition and removal can affect the chromatin at structural and functional levels. KDM5A is one of the four known H3K4-specific demethylases. It is a part of the KDM5 family, which is characterized by a catalytic Jumonji domain capable of removing H3K4 di- and tri-methylation marks. KDM5A has been found to be involved in multiple cellular processes such as differentiation, metabolism, cell cycle, and transcription. Its link to various diseases, including cancer, makes KDM5A an important target for drug development. However, despite several studies outlining its significance in various pathways, our lack of understanding of its recruitment and function at the target sites on the chromatin presents a challenge in creating effective and targeted treatments. Therefore, it is essential to understand the recruitment mechanism of KDM5A to chromatin, and its activity therein, to comprehend how various roles of KDM5A are regulated. In this review, we discuss how KDM5A functions in a context-dependent manner on the chromatin, either directly through its structural domain, or through various interacting partners, to bring about a diverse range of functions.
Collapse
Affiliation(s)
- Avishek Kataria
- Laboratory of Cell Cycle Regulation, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, India
- Graduate Studies, Manipal Academy of Higher Education, Manipal, India
| | - Shweta Tyagi
- Laboratory of Cell Cycle Regulation, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, India
| |
Collapse
|
5
|
Garg V, Kumar L. Metronomic chemotherapy in ovarian cancer. Cancer Lett 2023; 579:216469. [PMID: 37923056 DOI: 10.1016/j.canlet.2023.216469] [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: 09/04/2023] [Revised: 10/15/2023] [Accepted: 10/27/2023] [Indexed: 11/07/2023]
Abstract
Translational research and the development of targeted therapies have transformed the therapeutic landscape in epithelial ovarian cancer over the last decade. However, recurrent ovarian cancer continues to pose formidable challenges to therapeutic interventions, necessitating innovative strategies to optimize treatment outcomes. Current research focuses on the development of pharmaceuticals that target potential resistance pathways to DNA repair pathways. However, the cost and toxicity of some of these therapies are prohibitive and majority of patients lack access to clinical trials. Metronomic chemotherapy, characterized by the continuous administration of low doses of chemotherapeutic agents without long treatment breaks, has emerged as a promising approach with potential implications beyond recurrent setting. It acts primarily by inhibition of angiogenesis and activation of host immune system. We here review the mechanism of action of metronomic chemotherapy, as well as its current role, limitations, and avenues for further research in the management of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Vikas Garg
- Clinical Research Fellow, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, 700 University Avenue, 7th Floor, Station 7W386, M5G 1Z5, Toronto, ON, Canada.
| | - Lalit Kumar
- Oncology and BMT, Department of Medical Oncology, Artemis Hospital, Gurugram, India.
| |
Collapse
|
6
|
Bravetti G, Falvo P, Talarico G, Orecchioni S, Bertolini F. Metronomic chemotherapy, dampening of immunosuppressive cells, antigen presenting cell activation, and T cells. A quartet against refractoriness and resistance to checkpoint inhibitors. Cancer Lett 2023; 577:216441. [PMID: 37806515 DOI: 10.1016/j.canlet.2023.216441] [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: 08/03/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/10/2023]
Abstract
Chemotherapeutic agents have profound effects on cancer, stroma and immune cells that - in most cases - depend upon the dosage and schedule of administration. Preclinical and clinical studies summarized and discussed in the present review have demonstrated that maximum tolerable dosage (MTD) vs low-dosage, continuous (metronomic) administration of most chemotherapeutics have polarized effects on immune cells. In particular, metronomic schedules might be associated - among others effects - with activation of antigen presenting cells and generation of new T cell clones to enhance the activity of several types of immunotherapies. Ongoing and planned clinical trials in different types of cancer will confirm or dismiss this hypothesis and provide candidate biomarker data for the selection of patients who are likely to benefit from these combinatorial strategies.
Collapse
Affiliation(s)
- Giulia Bravetti
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20137, Milan, Italy; Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Paolo Falvo
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20137, Milan, Italy; Medical University of Vienna, (MUW), Borschkegasse 8A 1090, Wien, Austria
| | - Giovanna Talarico
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20137, Milan, Italy; Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Stefania Orecchioni
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20137, Milan, Italy; Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20137, Milan, Italy; Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy.
| |
Collapse
|
7
|
Mani N, Daiya A, Chowdhury R, Mukherjee S, Chowdhury S. Epigenetic adaptations in drug-tolerant tumor cells. Adv Cancer Res 2023; 158:293-335. [PMID: 36990535 DOI: 10.1016/bs.acr.2022.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Traditional chemotherapy against cancer is often severely hampered by acquired resistance to the drug. Epigenetic alterations and other mechanisms like drug efflux, drug metabolism, and engagement of survival pathways are crucial in evading drug pressure. Herein, growing evidence suggests that a subpopulation of tumor cells can often tolerate drug onslaught by entering a "persister" state with minimal proliferation. The molecular features of these persister cells are gradually unraveling. Notably, the "persisters" act as a cache of cells that can eventually re-populate the tumor post-withdrawal drug pressure and contribute to acquiring stable drug-resistant features. This underlines the clinical significance of the tolerant cells. Accumulating evidence highlights the importance of modulation of the epigenome as a critical adaptive strategy for evading drug pressure. Chromatin remodeling, altered DNA methylation, and de-regulation of non-coding RNA expression and function contribute significantly to this persister state. No wonder targeting adaptive epigenetic modifications is increasingly recognized as an appropriate therapeutic strategy to sensitize them and restore drug sensitivity. Furthermore, manipulating the tumor microenvironment and "drug holiday" is also explored to maneuver the epigenome. However, heterogeneity in adaptive strategies and lack of targeted therapies have significantly hindered the translation of epigenetic therapy to the clinics. In this review, we comprehensively analyze the epigenetic alterations adapted by the drug-tolerant cells, the therapeutic strategies employed to date, and their limitations and future prospects.
Collapse
|
8
|
JMJD family proteins in cancer and inflammation. Signal Transduct Target Ther 2022; 7:304. [PMID: 36050314 PMCID: PMC9434538 DOI: 10.1038/s41392-022-01145-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/22/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022] Open
Abstract
The occurrence of cancer entails a series of genetic mutations that favor uncontrollable tumor growth. It is believed that various factors collectively contribute to cancer, and there is no one single explanation for tumorigenesis. Epigenetic changes such as the dysregulation of enzymes modifying DNA or histones are actively involved in oncogenesis and inflammatory response. The methylation of lysine residues on histone proteins represents a class of post-translational modifications. The human Jumonji C domain-containing (JMJD) protein family consists of more than 30 members. The JMJD proteins have long been identified with histone lysine demethylases (KDM) and histone arginine demethylases activities and thus could function as epigenetic modulators in physiological processes and diseases. Importantly, growing evidence has demonstrated the aberrant expression of JMJD proteins in cancer and inflammatory diseases, which might serve as an underlying mechanism for the initiation and progression of such diseases. Here, we discuss the role of key JMJD proteins in cancer and inflammation, including the intensively studied histone lysine demethylases, as well as the understudied group of JMJD members. In particular, we focused on epigenetic changes induced by each JMJD member and summarized recent research progress evaluating their therapeutic potential for the treatment of cancer and inflammatory diseases.
Collapse
|
9
|
Tang DG. Understanding and targeting prostate cancer cell heterogeneity and plasticity. Semin Cancer Biol 2022; 82:68-93. [PMID: 34844845 PMCID: PMC9106849 DOI: 10.1016/j.semcancer.2021.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022]
Abstract
Prostate cancer (PCa) is a prevalent malignancy that occurs primarily in old males. Prostate tumors in different patients manifest significant inter-patient heterogeneity with respect to histo-morphological presentations and molecular architecture. An individual patient tumor also harbors genetically distinct clones in which PCa cells display intra-tumor heterogeneity in molecular features and phenotypic marker expression. This inherent PCa cell heterogeneity, e.g., in the expression of androgen receptor (AR), constitutes a barrier to the long-term therapeutic efficacy of AR-targeting therapies. Furthermore, tumor progression as well as therapeutic treatments induce PCa cell plasticity such that AR-positive PCa cells may turn into AR-negative cells and prostate tumors may switch lineage identity from adenocarcinomas to neuroendocrine-like tumors. This induced PCa cell plasticity similarly confers resistance to AR-targeting and other therapies. In this review, I first discuss PCa from the perspective of an abnormal organ development and deregulated cellular differentiation, and discuss the luminal progenitor cells as the likely cells of origin for PCa. I then focus on intrinsic PCa cell heterogeneity in treatment-naïve tumors with the presence of prostate cancer stem cells (PCSCs). I further elaborate on PCa cell plasticity induced by genetic alterations and therapeutic interventions, and present potential strategies to therapeutically tackle PCa cell heterogeneity and plasticity. My discussions will make it clear that, to achieve enduring clinical efficacy, both intrinsic PCa cell heterogeneity and induced PCa cell plasticity need to be targeted with novel combinatorial approaches.
Collapse
Affiliation(s)
- Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Experimental Therapeutics (ET) Graduate Program, The University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
10
|
Andrade da Mota TH, Reis Guimarães AF, Silva de Carvalho AÉ, Saldanha- de Araujo F, Pinto de Faria Lopes G, Pittella-Silva F, do Amaral Rabello D, Madureira de Oliveira D. Effects of in vitro short- and long-term treatment with telomerase inhibitor in U-251 glioma cells. Tumour Biol 2021; 43:327-340. [DOI: 10.3233/tub-211515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND: The inhibition of the enzyme telomerase (TERT) has been widely investigated as a new pharmacological approach for cancer treatment, but its real potential and the biochemical consequences are not totally understood. OBJECTIVE: Here, we investigated the effects of the telomerase inhibitor MST-312 on a human glioma cell line after both short- and long-term (290 days) treatments. METHODS: Effects on cell growth, viability, cell cycle, morphology, cell death and genes expression were assessed. RESULTS: We found that short-term treatment promoted cell cycle arrest followed by apoptosis. Importantly, cells with telomerase knock-down revealed that the toxic effects of MST-312 are partially TERT dependent. In contrast, although the long-term treatment decreased cell proliferation at first, it also caused adaptations potentially related to treatment resistance and tumor aggressiveness after long time of exposition. CONCLUSIONS: Despite the short-term effects of telomerase inhibition not being due to telomere erosion, they are at least partially related to the enzyme inhibition, which may represent an important strategy to pave the way for tumor growth control, especially through modulation of the non-canonical functions of telomerase. On the other hand, long-term exposure to the inhibitor had the potential to induce cell adaptations with possible negative clinical implications.
Collapse
Affiliation(s)
- Tales Henrique Andrade da Mota
- Multidisciplinary Laboratory of Human Health, University of Brasilia, Ceilândia, DF, Brazil
- Laboratory of Molecular Pathology of Cancer, University of Brasilia, Brasilia, DF, Brazil
| | - Ana Flávia Reis Guimarães
- Multidisciplinary Laboratory of Human Health, University of Brasilia, Ceilândia, DF, Brazil
- Laboratory of Molecular Pathology of Cancer, University of Brasilia, Brasilia, DF, Brazil
| | - Amandda Évelin Silva de Carvalho
- Laboratory of Molecular Pharmacology, Department of Pharmaceutical Sciences, University of Brasilia, Brasilia, DF, Brazil
- Laboratory of Hematology, Department of Pharmaceutical Sciences, University of Brasilia, Brasilia, DF, Brazil
| | - Felipe Saldanha- de Araujo
- Laboratory of Molecular Pharmacology, Department of Pharmaceutical Sciences, University of Brasilia, Brasilia, DF, Brazil
- Laboratory of Hematology, Department of Pharmaceutical Sciences, University of Brasilia, Brasilia, DF, Brazil
| | - Giselle Pinto de Faria Lopes
- Laboratory of Cellular and Molecular Hemato-oncology, National Institute of Cancer (INCA), Rio de Janeiro, RJ, Brazil
- Marine Biotechnology Department, Admiral Paulo Moreira Sea Studies Institute, IEAPM, Arraial do Cabo, RJ, Brazil
| | - Fábio Pittella-Silva
- Laboratory of Molecular Pathology of Cancer, University of Brasilia, Brasilia, DF, Brazil
| | | | - Diêgo Madureira de Oliveira
- Multidisciplinary Laboratory of Human Health, University of Brasilia, Ceilândia, DF, Brazil
- Laboratory of Molecular Pathology of Cancer, University of Brasilia, Brasilia, DF, Brazil
| |
Collapse
|
11
|
Yang GJ, Wu J, Miao L, Zhu MH, Zhou QJ, Lu XJ, Lu JF, Leung CH, Ma DL, Chen J. Pharmacological inhibition of KDM5A for cancer treatment. Eur J Med Chem 2021; 226:113855. [PMID: 34555614 DOI: 10.1016/j.ejmech.2021.113855] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/15/2022]
Abstract
Lysine-specific demethylase 5A (KDM5A, also named RBP2 or JARID1A) is a demethylase that can remove methyl groups from histones H3K4me1/2/3. It is aberrantly expressed in many cancers, where it impedes differentiation and contributes to cancer cell proliferation, cell metastasis and invasiveness, drug resistance, and is associated with poor prognosis. Pharmacological inhibition of KDM5A has been reported to significantly attenuate tumor progression in vitro and in vivo in a range of solid tumors and acute myeloid leukemia. This review will present the structural aspects of KDM5A, its role in carcinogenesis, a comparison of currently available approaches for screening KDM5A inhibitors, a classification of KDM5A inhibitors, and its potential as a drug target in cancer therapy.
Collapse
Affiliation(s)
- Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Jia Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, 999078, China
| | - Liang Miao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Ming-Hui Zhu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Qian-Jin Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Xin-Jiang Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Jian-Fei Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, 999078, China.
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong, 999077, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
12
|
Picon H, Guddati AK. Cancer stem cells in head and neck cancer. AMERICAN JOURNAL OF STEM CELLS 2021; 10:28-35. [PMID: 34552815 PMCID: PMC8449141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/15/2021] [Indexed: 06/13/2023]
Abstract
Cancer stem cells (CSCs) are a unique population of cells found within tumors that are able to self-renew, restore the original heterogeneity of a tumor following treatment, and show increased tumorigenic potential when compared to other cancer cells. It is thought that they are responsible for the recurrence of tumors as well as the resistance to treatment that is seen clinically. CSCs are known to be involved in head and neck cancer (HNCs) specifically, as evidence for their existence can be found in head and neck squamous cell carcinoma (HNSCC), mucoepidermoid carcinoma (MEC), and adenoid cystic carcinoma (ACC), among others. Here, findings from various approaches to identifying and targeting CSCs and their downstream effectors in HNC are summarized, with an emphasis on recent advancements. Prognostic and therapeutic markers are discussed for each specific type of HNC, and novel treatment strategies and current clinical trials involving CSCs are detailed as well. The information provided here is intended to further the research on this important topic and lead to clinical impact in the battle against HNC.
Collapse
Affiliation(s)
- Hector Picon
- Medical College of Georgia, Augusta UniversityAugusta 30909, GA, USA
| | - Achuta Kumar Guddati
- Division of Hematology/Oncology, Georgia Cancer Center, Augusta UniversityAugusta 30912, GA, USA
| |
Collapse
|
13
|
Yang GJ, Zhu MH, Lu XJ, Liu YJ, Lu JF, Leung CH, Ma DL, Chen J. The emerging role of KDM5A in human cancer. J Hematol Oncol 2021; 14:30. [PMID: 33596982 PMCID: PMC7888121 DOI: 10.1186/s13045-021-01041-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Histone methylation is a key posttranslational modification of chromatin, and its dysregulation affects a wide array of nuclear activities including the maintenance of genome integrity, transcriptional regulation, and epigenetic inheritance. Variations in the pattern of histone methylation influence both physiological and pathological events. Lysine-specific demethylase 5A (KDM5A, also known as JARID1A or RBP2) is a KDM5 Jumonji histone demethylase subfamily member that erases di- and tri-methyl groups from lysine 4 of histone H3. Emerging studies indicate that KDM5A is responsible for driving multiple human diseases, particularly cancers. In this review, we summarize the roles of KDM5A in human cancers, survey the field of KDM5A inhibitors including their anticancer activity and modes of action, and the current challenges and potential opportunities of this field.
Collapse
Affiliation(s)
- Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, People's Republic of China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China.,Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, People's Republic of China
| | - Ming-Hui Zhu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, People's Republic of China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Xin-Jiang Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, People's Republic of China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Yan-Jun Liu
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, 210046, People's Republic of China
| | - Jian-Fei Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, People's Republic of China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Chung-Hang Leung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, People's Republic of China.
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong, 999077, People's Republic of China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China. .,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, People's Republic of China. .,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China.
| |
Collapse
|
14
|
Paik S, Maule F, Gallo M. Dysregulation of chromatin organization in pediatric and adult brain tumors: oncoepigenomic contributions to tumorigenesis and cancer stem cell properties. Genome 2020; 64:326-336. [PMID: 33075237 DOI: 10.1139/gen-2020-0097] [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: 11/22/2022]
Abstract
The three-dimensional (3D) organization of the genome is a crucial enabler of cell fate, identity, and function. In this review, we will focus on the emerging role of altered 3D genome organization in the etiology of disease, with a special emphasis on brain cancers. We discuss how different genetic alterations can converge to disrupt the epigenome in childhood and adult brain tumors, by causing aberrant DNA methylation and by affecting the amounts and genomic distribution of histone post-translational modifications. We also highlight examples that illustrate how epigenomic alterations have the potential to affect 3D genome architecture in brain tumors. Finally, we will propose the concept of "epigenomic erosion" to explain the transition from stem-like cells to differentiated cells in hierarchically organized brain cancers.
Collapse
Affiliation(s)
- Seungil Paik
- Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Francesca Maule
- Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Marco Gallo
- Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
15
|
Shu Y, Weng S, Zheng S. Metronomic chemotherapy in non-small cell lung cancer. Oncol Lett 2020; 20:307. [PMID: 33093916 DOI: 10.3892/ol.2020.12170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 07/15/2020] [Indexed: 12/17/2022] Open
Abstract
Metronomic chemotherapy (MCT) is defined as the rhythmic chemotherapy of low-dose cytotoxic drugs with short or no drug-free breaks over prolonged periods. MCT affects tumor cells and the tumor microenvironment. Particularly, the low-dose schedule impairs the repair process of endothelial cells, resulting in an anti-angiogenesis effect. By stimulating the immune system to eliminate tumor cells, MCT induces immunological activation. Furthermore, combined with targeted therapy, anti-angiogenic drugs enhance the efficacy of MCT. The present review is an overview of phase I, II and III clinical trials focusing on the efficacy, toxicity and mechanism of MCT in patients with non-small cell lung cancer (NSCLC). Furthermore, the prospects of MCT in NSCLC have been discussed. The present review indicated that MCT is an efficacious treatment for selected patients with NSCLC, with acceptable systemic side effects and economic viability for public health.
Collapse
Affiliation(s)
- Yefei Shu
- Department of Medical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China
| | - Shanshan Weng
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Song Zheng
- Department of Medical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, P.R. China.,Department of Medical Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
16
|
Targeting of Histone Demethylases KDM5A and KDM6B Inhibits the Proliferation of Temozolomide-Resistant Glioblastoma Cells. Cancers (Basel) 2019; 11:cancers11060878. [PMID: 31238504 PMCID: PMC6627323 DOI: 10.3390/cancers11060878] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/03/2019] [Accepted: 06/17/2019] [Indexed: 01/25/2023] Open
Abstract
Lysine histone demethylases (KDMs) are considered potential therapeutic targets in several tumors, including glioblastoma (GB). In particular, KDM5A is involved in the acquisition of temozolomide (TMZ) resistance in adult GB cells and UDX/KDM6B regulates H3K27 methylation, which is involved in the pediatric diffuse intrinsic pontine glioma (DIPG). Synthetic inhibitors of KDM5A (JIB 04 and CPI-455) efficiently block the proliferation of native and TMZ-resistant cells and the KDM6B inhibitor GSK J4 improves survival in a model of DIPG. The aim of our work was to determine if GSK J4 could be effective against GB cells that have acquired TMZ resistance and if it could synergize with TMZ or JIB 04 to increase the clinical utility of these molecules. Standard functional and pharmacological analytical procedures were utilized to determine the efficacy of the molecules under study when used alone or in combination against native GB cells and in a model of drug resistance. The results of this study indicated that although GSK J4 is active against native and TMZ-resistant cells, it does so at a lower efficacy than JIB 04. Drug combination studies revealed that GSK J4, differently from JIB 04, does not synergize with TMZ. Interestingly, GSK J4 and JIB 04 strongly synergize and are a potent combination against TMZ-resistant cells. Further studies in animal models will be necessary to determine if this combination of molecules might foster the development of novel therapeutic approaches for glioblastoma.
Collapse
|
17
|
Simsek C, Esin E, Yalcin S. Metronomic Chemotherapy: A Systematic Review of the Literature and Clinical Experience. JOURNAL OF ONCOLOGY 2019; 2019:5483791. [PMID: 31015835 PMCID: PMC6446118 DOI: 10.1155/2019/5483791] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 12/24/2018] [Accepted: 02/05/2019] [Indexed: 02/07/2023]
Abstract
Metronomic chemotherapy, continuous and dose-dense administration of chemotherapeutic drugs with lowered doses, is being evaluated for substituting, augmenting, or appending conventional maximum tolerated dose regimens, with preclinical and clinical studies for the past few decades. To date, the principle mechanisms of its action include impeding tumoral angiogenesis and modulation of hosts' immune system, affecting directly tumor cells, their progenitors, and neighboring stromal cells. Its better toxicity profile, lower cost, and easier use are main advantages over conventional therapies. The evidence of metronomic chemotherapy for personalized medicine is growing, starting with unfit elderly patients and also for palliative treatment. The literature reviewed in this article mainly demonstrates that metronomic chemotherapy is advantageous for selected patients and for certain types of malignancies, which make it a promising therapeutic approach for filling in the gaps. More clinical studies are needed to establish a solidified role for metronomic chemotherapy with other treatment models in modern cancer management.
Collapse
Affiliation(s)
- Cem Simsek
- Department of Internal Medicine, Hacettepe University, Ankara, Turkey
| | - Ece Esin
- Department of Medical Oncology, A.Y. Ankara Training Hospital, Ankara, Turkey
| | - Suayib Yalcin
- Department of Medical Oncology, Hacettepe University, Ankara, Turkey
| |
Collapse
|
18
|
Romani M, Pistillo MP, Banelli B. Epigenetic Targeting of Glioblastoma. Front Oncol 2018; 8:448. [PMID: 30386738 PMCID: PMC6198064 DOI: 10.3389/fonc.2018.00448] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/24/2018] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma is one of the first tumors where the biological changes accompanying a single epigenetic modification, the methylation of the MGMT gene, were found to be of clinical relevance. The exploration of the epigenomic landscape of glioblastoma has allowed to identify patients carrying a diffuse hypermethylation at gene promoters and with better outcome. Epigenetic and genetic data have led to the definition of major subgroups of glioma and were the basis of the current WHO classification of CNS tumors and of a novel classification based solely on DNA methylation data that shows a remarkable diagnostic precision.The reversibility of epigenetic modifications is considered a therapeutic opportunity in many tumors also because these alterations have been mechanistically linked to the biological characteristics of glioblastoma. Several alterations like IDH1/2 mutations that interfere with "epigenetic modifier" enzymes, the mutations of the histone 3 variants H3.1 and H3.3 that alter the global H3K27me3 levels and the altered expression of histone methyltransferases and demethylases are considered potentially druggable targets in glioma and molecules targeting these alterations are being tested in preclinical and clinical trials. The recent advances on the knowledge of the players of the "epigenetic orchestra" and of their mutual interactions are indicating new paths that may eventually open new therapeutic options for this invariably lethal cancer.
Collapse
Affiliation(s)
- Massimo Romani
- Laboratory of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Maria Pia Pistillo
- Laboratory of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Barbara Banelli
- Laboratory of Tumor Epigenetics, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Health Sciences, University of Genoa, Genova, Italy
| |
Collapse
|
19
|
Banelli B, Daga A, Forlani A, Allemanni G, Marubbi D, Pistillo MP, Profumo A, Romani M. Small molecules targeting histone demethylase genes (KDMs) inhibit growth of temozolomide-resistant glioblastoma cells. Oncotarget 2018; 8:34896-34910. [PMID: 28432280 PMCID: PMC5471020 DOI: 10.18632/oncotarget.16820] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 03/22/2017] [Indexed: 12/29/2022] Open
Abstract
In glioblastoma several histone demethylase genes (KDM) are overexpressed compared to normal brain tissue and the development of Temozolomide (TMZ) resistance is accompanied by the transient further increased expression of KDM5A and other KDMs following a mechanism that we defined as “epigenetic resilience”. We hypothesized that targeting KDMs may kill the cells that survive the cytotoxic therapy. We determined the effect of JIB 04 and CPI-455, two KDM inhibitors, on glioblastoma cells and found that both molecules are more effective against TMZ-resistant rather than native cells. Because of its lower IC50, we focused on JIB 04 that targets KDM5A and other KDMs as well. We have shown that this molecule activates autophagic and apoptotic pathways, interferes with cell cycle progression, inhibits cell clonogenicity and dephosphorylates Akt thus inactivating a potent pro-survival pathway. We performed combination temozolomide/JIB 04 in vitro treatments showing that these two molecules, under certain conditions, have a strong synergic effect and we hypothesize that JIB 04 intercepts the cells that escape the G2 block exerted by TMZ. Finally we studied the permeability of JIB 04 across the blood-brain barrier and found that this molecule reaches bioactive concentration in the brain; furthermore a pilot in vivo experiment in an orthotopic GB xenograft model showed a trend toward longer survival in treated mice with an Hazard Ratio of 0.5. In conclusion we propose that the combination between cytotoxic drugs and molecules acting on the epigenetic landscape may offer the opportunity to develop new therapies for this invariably lethal disease.
Collapse
Affiliation(s)
- Barbara Banelli
- Laboratory of Tumor Epigenetics, IRCCS AOU San Martino-IST, Genova, Italy.,Department of Health Sciences, University of Genova, Genova, Italy
| | - Antonio Daga
- Laboratory of Regenerative Medicine, IRCCS AOU San Martino-IST, Genova, Italy
| | - Alessandra Forlani
- Laboratory of Tumor Epigenetics, IRCCS AOU San Martino-IST, Genova, Italy
| | - Giorgio Allemanni
- Laboratory of Tumor Epigenetics, IRCCS AOU San Martino-IST, Genova, Italy
| | - Daniela Marubbi
- Laboratory of Regenerative Medicine, IRCCS AOU San Martino-IST, Genova, Italy.,Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Maria Pia Pistillo
- Laboratory of Tumor Epigenetics, IRCCS AOU San Martino-IST, Genova, Italy
| | - Aldo Profumo
- Biopolymers and Proteomic Unit, IRCCS AOU San Martino-IST, Genova, Italy
| | - Massimo Romani
- Laboratory of Tumor Epigenetics, IRCCS AOU San Martino-IST, Genova, Italy
| |
Collapse
|
20
|
Lin HM, Lee BY, Castillo L, Spielman C, Grogan J, Yeung NK, Kench JG, Stricker PD, Haynes AM, Centenera MM, Butler LM, Shreeve SM, Horvath LG, Daly RJ. Effect of FAK inhibitor VS-6063 (defactinib) on docetaxel efficacy in prostate cancer. Prostate 2018; 78:308-317. [PMID: 29314097 DOI: 10.1002/pros.23476] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/13/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Docetaxel, the standard chemotherapy for metastatic castration-resistant prostate cancer (CRPC) also enhances the survival of patients with metastatic castration-sensitive prostate cancer (CSPC) when combined with androgen-deprivation therapy. Focal Adhesion Kinase (FAK) activation is a mediator of docetaxel resistance in prostate cancer cells. The aim of this study was to investigate the effect of the second generation FAK inhibitor VS-6063 on docetaxel efficacy in pre-clinical CRPC and CSPC models. METHODS Docetaxel-resistant CRPC cells, mice with PC3 xenografts, and ex vivo cultures of patient-derived primary prostate tumors were treated with VS-6063 and/or docetaxel, or vehicle control. Cell counting, immunoblotting, and immunohistochemistry techniques were used to evaluate the treatment effects. RESULTS Docetaxel and VS-6063 co-treatment caused a greater decrease in the viability of docetaxel-resistant CRPC cells, and a greater inhibition in PC3 xenograft growth compared to either monotherapy. FAK expression in human primary prostate cancer was positively associated with advanced tumor stage. Patient-derived prostate tumor explants cultured with both docetaxel and VS-6063 displayed a higher percentage of apoptosis in cancer cells, than monotherapy treatment. CONCLUSIONS Our findings suggest that co-administration of the FAK inhibitor, VS-6063, with docetaxel represents a potential therapeutic strategy to overcome docetaxel resistance in prostate cancer.
Collapse
Affiliation(s)
- Hui-Ming Lin
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St Vincent's Clinical School, The University of New South Wales, Darlinghurst, New South Wales, Australia
| | - Brian Y Lee
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Lesley Castillo
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Calan Spielman
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Judith Grogan
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Nicole K Yeung
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - James G Kench
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Australian Prostate Cancer Research Centre-NSW, Darlinghurst, New South Wales, Australia
| | - Phillip D Stricker
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St Vincent's Clinical School, The University of New South Wales, Darlinghurst, New South Wales, Australia
- Australian Prostate Cancer Research Centre-NSW, Darlinghurst, New South Wales, Australia
- St Vincent's Prostate Cancer Centre, Darlinghurst, New South Wales, Australia
| | - Anne-Maree Haynes
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Australian Prostate Cancer Research Centre-NSW, Darlinghurst, New South Wales, Australia
| | - Margaret M Centenera
- School of Medicine and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Lisa M Butler
- School of Medicine and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - S Martin Shreeve
- Janssen Pharmaceutical Companies of Johnson and Johnson, San Diego, California
| | - Lisa G Horvath
- Cancer Division, The Kinghorn Cancer Centre/Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Australian Prostate Cancer Research Centre-NSW, Darlinghurst, New South Wales, Australia
- Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | - Roger J Daly
- Signalling Network Laboratory, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
21
|
André N, Tsai K, Carré M, Pasquier E. Metronomic Chemotherapy: Direct Targeting of Cancer Cells after all? Trends Cancer 2017; 3:319-325. [DOI: 10.1016/j.trecan.2017.03.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/25/2017] [Accepted: 03/29/2017] [Indexed: 12/22/2022]
|
22
|
Ahmed M, Chaudhari K, Babaei-Jadidi R, Dekker LV, Shams Nateri A. Concise Review: Emerging Drugs Targeting Epithelial Cancer Stem-Like Cells. Stem Cells 2017; 35:839-850. [DOI: 10.1002/stem.2579] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 01/03/2017] [Accepted: 01/07/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Mehreen Ahmed
- Cancer Genetics & Stem Cell Group; Nottingham United Kingdom
| | | | - Roya Babaei-Jadidi
- Cancer Genetics & Stem Cell Group; Nottingham United Kingdom
- Tumor & Vascular Biology Laboratories; Cancer Biology, Division of Cancer and Stem Cells, School of Medicine; Nottingham United Kingdom
| | - Lodewijk V. Dekker
- Division of Medicinal Chemistry and Structural Biology, School of Pharmacy; Centre for Biomolecular Science, University of Nottingham; Nottingham United Kingdom
| | | |
Collapse
|
23
|
Biziota E, Mavroeidis L, Hatzimichael E, Pappas P. Metronomic chemotherapy: A potent macerator of cancer by inducing angiogenesis suppression and antitumor immune activation. Cancer Lett 2016; 400:243-251. [PMID: 28017892 DOI: 10.1016/j.canlet.2016.12.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/14/2016] [Accepted: 12/15/2016] [Indexed: 01/09/2023]
Abstract
Metronomic chemotherapy is a low dosing treatment strategy that attracts growing scientific and clinical interest. It refers to dense and uninterrupted administration of low doses of chemotherapeutic agents (without prolonged drug free intervals) over extended periods of time. Cancer chemotherapy is conventionally given in cycles of maximum tolerated doses (MTD) with the aim of inducing maximum cancer cell apoptosis. In contrast, the primary target of metronomic chemotherapy is the tumor's neovasculature. This is relevant to the emerging concept that tumors exist in a complex microenvironment of cancer cells, stromal cells and supporting vessels. In addition to its anti-angiogenetic properties, metronomic chemotherapy halts tumor growth by activating anti-tumor immunity, thus decreasing the acquired resistance to conventional chemotherapy. Herein, we present a review of the literature that provides a scientific basis for the merits of chemotherapy when administered on a metronomic schedule.
Collapse
Affiliation(s)
- Eirini Biziota
- Department of Medical Oncology, University Hospital of Evros, Alexandroupolis, 68 100, Greece.
| | - Leonidas Mavroeidis
- Department of Pharmacology, Faculty of Medicine, School of Life Sciences, University of Ioannina, Ioannina, 451 10, Greece.
| | | | - Periklis Pappas
- Department of Pharmacology, Faculty of Medicine, School of Life Sciences, University of Ioannina, Ioannina, 451 10, Greece.
| |
Collapse
|
24
|
Banelli B, Carra E, Barbieri F, Würth R, Parodi F, Pattarozzi A, Carosio R, Forlani A, Allemanni G, Marubbi D, Florio T, Daga A, Romani M. The histone demethylase KDM5A is a key factor for the resistance to temozolomide in glioblastoma. Cell Cycle 2016; 14:3418-29. [PMID: 26566863 DOI: 10.1080/15384101.2015.1090063] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Notwithstanding current multimodal treatment, including surgery, radiotherapy and chemotherapy with temozolomide (TMZ), median survival of glioblastoma (GBM) patients is about 14 months, due to the rapid emergence of cell clones resistant to treatment. Therefore, understanding the mechanisms underlying chemoresistance is mandatory to improve treatments' outcome. We generated TMZ resistant cells (TMZ-R) from a GBM cell line and from cancer stem cell-enriched cultures isolated from human GBMs. We demonstrated that TMZ resistance is partially reverted by "drug wash-out" suggesting the contribution of epigenetic mechanisms in drug resistance and supporting the possibility of TMZ rechallenge in GBM patients after prior drug exposure. The expression of histone lysine demethylase genes (KDMs) was increased in TMZ-R cells compared to parental cells, and TMZ resistance or restored sensitivity was mimicked by over-expressing or inactivating KDM5A. Methylation and expression of O6-methylguanine-DNA methyltransferase (MGMT) and drug efflux mechanisms were not altered in TMZ-R cells compared to parental TMZ sensitive cells. TMZ-R cells transiently acquired morphologic and molecular characteristics of differentiated tumor cells, features that were lost after drug wash-out. In conclusion, we demonstrated that treatment-induced TMZ resistance in GBM involves epigenetic mechanisms in a subset of slow-cycling and transiently partially differentiated cells that escape drug cytotoxicity, overcome G2 checkpoint and sustain clonal growth. We found that TMZ-R cells are sensitive to histone deacethylase inhibitors (HDACi) that synergize with TMZ. This strong synergism could be exploited to develop novel combined adjuvant therapies for this rapidly progressing and invariably lethal cancer.
Collapse
Affiliation(s)
- Barbara Banelli
- a Laboratory of Tumor Epigenetics; IRCCS AOU San Martino - IST ; Genova , Italy
| | - Elisa Carra
- b Laboratory of Regenerative Medicine; IRCCS AOU San Martino - IST ; Genova , Italy
| | - Federica Barbieri
- c Section of Pharmacology ; Department of Internal Medicine and Center of Excellence for Biomedical Research (CEBR); University of Genova ; Genova , Italy
| | - Roberto Würth
- c Section of Pharmacology ; Department of Internal Medicine and Center of Excellence for Biomedical Research (CEBR); University of Genova ; Genova , Italy
| | - Federica Parodi
- a Laboratory of Tumor Epigenetics; IRCCS AOU San Martino - IST ; Genova , Italy
| | - Alessandra Pattarozzi
- c Section of Pharmacology ; Department of Internal Medicine and Center of Excellence for Biomedical Research (CEBR); University of Genova ; Genova , Italy
| | - Roberta Carosio
- a Laboratory of Tumor Epigenetics; IRCCS AOU San Martino - IST ; Genova , Italy
| | - Alessandra Forlani
- a Laboratory of Tumor Epigenetics; IRCCS AOU San Martino - IST ; Genova , Italy
| | - Giorgio Allemanni
- a Laboratory of Tumor Epigenetics; IRCCS AOU San Martino - IST ; Genova , Italy
| | - Daniela Marubbi
- b Laboratory of Regenerative Medicine; IRCCS AOU San Martino - IST ; Genova , Italy.,d Department of Experimental Medicine (DIMES) ; University of Genova ; Genova , Italy
| | - Tullio Florio
- c Section of Pharmacology ; Department of Internal Medicine and Center of Excellence for Biomedical Research (CEBR); University of Genova ; Genova , Italy
| | - Antonio Daga
- b Laboratory of Regenerative Medicine; IRCCS AOU San Martino - IST ; Genova , Italy
| | - Massimo Romani
- a Laboratory of Tumor Epigenetics; IRCCS AOU San Martino - IST ; Genova , Italy
| |
Collapse
|
25
|
Koukourakis MI, Kakouratos C, Kalamida D, Bampali Z, Mavropoulou S, Sivridis E, Giatromanolaki A. Hypoxia-inducible proteins HIF1α and lactate dehydrogenase LDH5, key markers of anaerobic metabolism, relate with stem cell markers and poor post-radiotherapy outcome in bladder cancer. Int J Radiat Biol 2016; 92:353-63. [PMID: 27010533 DOI: 10.3109/09553002.2016.1162921] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE To assess whether anaerobic metabolism, proliferation activity and stem cell content are linked with radioresistance in bladder cancer. MATERIALS AND METHODS Tissue sections from 66 patients with invasive transitional cell bladder cancer treated with hypofractionated accelerated radiotherapy, was immunohistochemically analyzed for the Hypoxia-Inducible Factor 1α (HIF1α) and the anaerobic glycolysis enzyme lactate dehydrogenase 5 (LDH5). Proliferation index (Ki-67) and stem-cell marker (cluster of differentiation CD44, aldehyde dehydrogenase ALDH1) expression was also examined. RESULTS Both HIF1α and LDH5 expression were linked with high CD44 stem cell population (p = 0.001 and 0.05, respectively), while high Ki-67 proliferation index was linked with nuclear LDH5 expression (p = 0.03) and high histological grade (p = 0.02). A strong significant association of HIF1α (p = 0.0009) and of LDH5 (p < 0.0001) with poor local relapse free survival (LRFS) was noted, which was also confirmed in multivariate analysis. A significant association with overall survival was also noted. Silencing of lactate dehydrogenase LDHA gene in the human RT112 bladder cancer cell line, or exposure to oxamate (LDH activity inhibitor), resulted in strong radio-sensitization. CONCLUSIONS HIF1α and LDH5 are markers of poor outcome in patients with bladder cancer treated with radiotherapy. Blockage of anaerobic metabolism may prove of importance in clinical radiotherapy.
Collapse
Affiliation(s)
- Michael I Koukourakis
- a Department of Radiotherapy/Oncology , Democritus University of Thrace, and University General Hospital of Alexandroupolis , Alexandroupolis , Greece
| | - Christos Kakouratos
- a Department of Radiotherapy/Oncology , Democritus University of Thrace, and University General Hospital of Alexandroupolis , Alexandroupolis , Greece
| | - Dimitra Kalamida
- a Department of Radiotherapy/Oncology , Democritus University of Thrace, and University General Hospital of Alexandroupolis , Alexandroupolis , Greece
| | - Zoi Bampali
- a Department of Radiotherapy/Oncology , Democritus University of Thrace, and University General Hospital of Alexandroupolis , Alexandroupolis , Greece
| | | | - Efthimios Sivridis
- b Department of Pathology , Democritus University of Thrace, and University General Hospital of Alexandroupolis , Alexandroupolis , Greece
| | - Alexandra Giatromanolaki
- b Department of Pathology , Democritus University of Thrace, and University General Hospital of Alexandroupolis , Alexandroupolis , Greece
| |
Collapse
|
26
|
Zhang W, Meng Y, Liu N, Wen XF, Yang T. Insights into Chemoresistance of Prostate Cancer. Int J Biol Sci 2015; 11:1160-70. [PMID: 26327810 PMCID: PMC4551752 DOI: 10.7150/ijbs.11439] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 06/29/2015] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer (PCa) remains the most prevalent malignancy among males in the western world. Though hormonal therapies through chemical or surgical castration have been proposed many years ago, heretofore, such mainstay for the treatment on advanced PCa has not fundamentally changed. These therapeutic responses are temporary and most cases will eventually undergo PCa recurrence and metastasis, or even progress to castration-resistant prostate cancer (CRPC) due to persistent development of drug resistance. Prostate cancer stem cells (PCSCs) are a small population of cells, which possess unlimited self-renewal capacities, and can regenerate tumorigenic progenies, and play an essential role in PCa therapy resistance, metastasis and recurrence. Nowadays advanced progresses have been made in understanding of PCSC properties, roles of androgen receptor signaling and ATP-binding cassette sub-family G member 2 (ABCG2), as well as roles of genomic non-coding microRNAs and key signaling pathways, which have led to the development of novel therapies which are active against chemoresistant PCa and CRPC. Based on these progresses, this review is dedicated to address mechanisms underlying PCa chemoresistance, unveil crosstalks among pivotal signaling pathways, explore novel biotherapeutic agents, and elaborate functional properties and specific roles of chemoresistant PCSCs, which may act as a promising target for novel therapies against chemoresistant PCa.
Collapse
Affiliation(s)
- Wei Zhang
- 1. Department of Pharmacology, School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yan Meng
- 2. Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Na Liu
- 3. Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiao-Fei Wen
- 4. Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Tao Yang
- 2. Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
27
|
Zhang S, Liu J, Cheng Y. [Metronomic Chemotherapy--A New Path to Treat Advanced Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 18:232-9. [PMID: 25936888 PMCID: PMC6000289 DOI: 10.3779/j.issn.1009-3419.2015.04.08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
节拍化疗是近年来兴起的一种新的化疗策略。与传统化疗不同,节拍化疗通过相对低剂量的、频繁的应用细胞毒性药物,没有较长的治疗间歇,发挥抗肿瘤作用。最初认为节拍化疗直接作用于肿瘤血管内皮细胞,发挥抗血管生成作用。近年来发现节拍化疗还有调节机体免疫功能,影响肿瘤干细胞,诱导细胞休眠的作用。晚期非小细胞肺癌(non-small cell lung cancer, NSCLC)的治疗已经从彻底的清除肿瘤细胞转向改善疗效、降低毒性和提高生活质量。节拍化疗可以避免传统化疗毒性大,作用不持久的缺点,目前一些临床研究正在探索节拍化疗对晚期NSCLC的作用,并且初见疗效,有望成为晚期NSCLC一种新的治疗模式。
Collapse
Affiliation(s)
- Shuang Zhang
- Department of Thoracic Oncology, Jilin Provincial Cancer Hospital, Changchun 130012, China
| | - Jingjing Liu
- Department of Thoracic Oncology, Jilin Provincial Cancer Hospital, Changchun 130012, China
| | - Ying Cheng
- Department of Thoracic Oncology, Jilin Provincial Cancer Hospital, Changchun 130012, China
| |
Collapse
|
28
|
Yang T, Rycaj K. Targeted therapy against cancer stem cells. Oncol Lett 2015; 10:27-33. [PMID: 26170972 DOI: 10.3892/ol.2015.3172] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 04/21/2015] [Indexed: 01/05/2023] Open
Abstract
Research into cancer stem cells (CSCs), which have the ability to self-renew and give rise to more mature (differentiated) cancer cells, and which may be the cells responsible for the overall organization of a tumor, has progressed rapidly and concomitantly with recent advances in studies of normal tissue stem cells. CSCs have been reported in a wide spectrum of human tumors. Like normal tissue stem cells, CSCs similarly exhibit significant phenotypic and functional heterogeneity. The ability of CSCs to self-renew results in the immortality of malignant cells at the population level, whereas the ability of CSCs to differentiate, either fully or partially, generates the cellular hierarchy and heterogeneity commonly observed in solid tumors. CSCs also appear to have maximized their pro-survival mechanisms leading to their relative resistance to anti-cancer therapies and subsequent relapse. Studies in animal models of human cancers have also provided insight into the heterogeneity and characteristics of CSCs, helping to establish a platform for the development of novel targeted therapies against specific CSCs. In the present study, we briefly review the most recent progress in dissecting CSC heterogeneity and targeting CSCs in various human tumor systems. We also highlight a few examples of CSC-targeted drug development and clinical trials, with the ultimate aim of developing more effective therapeutic regimens that are capable of preventing tumor recurrence and metastasis.
Collapse
Affiliation(s)
- Tao Yang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Kiera Rycaj
- Department of Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| |
Collapse
|
29
|
Epithelial plasticity in prostate cancer: principles and clinical perspectives. Trends Mol Med 2014; 20:643-51. [PMID: 25262538 DOI: 10.1016/j.molmed.2014.09.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 01/23/2023]
Abstract
Over the past decade, the capacity of cancer cells to oscillate between epithelial and mesenchymal phenotypes, termed epithelial plasticity (EP), has been demonstrated to play a critical role in metastasis. This phenomenon may be particularly important for prostate cancer (PC) progression, since recent studies have revealed interplay between EP and signaling by the androgen receptor (AR) oncoprotein. Moreover, EP appears to play a role in dictating the response to therapies for metastatic PC. This review will evaluate preclinical and clinical evidence for the relevance of EP in PC progression and consider the potential of targeting and measuring EP as a means to treat and manage lethal forms of the disease.
Collapse
|
30
|
Bitting RL, Schaeffer D, Somarelli JA, Garcia-Blanco MA, Armstrong AJ. The role of epithelial plasticity in prostate cancer dissemination and treatment resistance. Cancer Metastasis Rev 2014; 33:441-68. [PMID: 24414193 PMCID: PMC4230790 DOI: 10.1007/s10555-013-9483-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nearly 30,000 men die annually in the USA of prostate cancer, nearly uniformly from metastatic dissemination. Despite recent advances in hormonal, immunologic, bone-targeted, and cytotoxic chemotherapies, treatment resistance and further dissemination are inevitable in men with metastatic disease. Emerging data suggests that the phenomenon of epithelial plasticity, encompassing both reversible mesenchymal transitions and acquisition of stemness traits, may underlie this lethal biology of dissemination and treatment resistance. Understanding the molecular underpinnings of this cellular plasticity from preclinical models of prostate cancer and from biomarker studies of human metastatic prostate cancer has provided clues to novel therapeutic approaches that may delay or prevent metastatic disease and lethality over time. This review will discuss the preclinical and clinical evidence for epithelial plasticity in this rapidly changing field and relate this to clinical phenotype and resistance in prostate cancer while suggesting novel therapeutic approaches.
Collapse
Affiliation(s)
- Rhonda L. Bitting
- Division of Medical Oncology, Duke Cancer Institute, Duke University, DUMC Box 102002, Durham, NC 27710, USA. Department of Medicine, Duke University, Durham, NC, USA. Center for RNA Biology, Duke University, Durham, NC, USA
| | - Daneen Schaeffer
- Center for RNA Biology, Duke University, Durham, NC, USA. Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Jason A. Somarelli
- Center for RNA Biology, Duke University, Durham, NC, USA. Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Mariano A. Garcia-Blanco
- Department of Medicine, Duke University, Durham, NC, USA. Center for RNA Biology, Duke University, Durham, NC, USA. Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Andrew J. Armstrong
- Division of Medical Oncology, Duke Cancer Institute, Duke University, DUMC Box 102002, Durham, NC 27710, USA. Department of Medicine, Duke University, Durham, NC, USA. Center for RNA Biology, Duke University, Durham, NC, USA. Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| |
Collapse
|
31
|
Phenotypic screening in cancer drug discovery - past, present and future. Nat Rev Drug Discov 2014; 13:588-602. [PMID: 25033736 DOI: 10.1038/nrd4366] [Citation(s) in RCA: 318] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There has been a resurgence of interest in the use of phenotypic screens in drug discovery as an alternative to target-focused approaches. Given that oncology is currently the most active therapeutic area, and also one in which target-focused approaches have been particularly prominent in the past two decades, we investigated the contribution of phenotypic assays to oncology drug discovery by analysing the origins of all new small-molecule cancer drugs approved by the US Food and Drug Administration (FDA) over the past 15 years and those currently in clinical development. Although the majority of these drugs originated from target-based discovery, we identified a significant number whose discovery depended on phenotypic screening approaches. We postulate that the contribution of phenotypic screening to cancer drug discovery has been hampered by a reliance on 'classical' nonspecific drug effects such as cytotoxicity and mitotic arrest, exacerbated by a paucity of mechanistically defined cellular models for therapeutically translatable cancer phenotypes. However, technical and biological advances that enable such mechanistically informed phenotypic models have the potential to empower phenotypic drug discovery in oncology.
Collapse
|
32
|
André N, Carré M, Pasquier E. Metronomics: towards personalized chemotherapy? Nat Rev Clin Oncol 2014; 11:413-31. [PMID: 24913374 DOI: 10.1038/nrclinonc.2014.89] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Since its inception in 2000, metronomic chemotherapy has undergone major advances as an antiangiogenic therapy. The discovery of the pro-immune properties of chemotherapy and its direct effects on cancer cells has established the intrinsic multitargeted nature of this therapeutic approach. The past 10 years have seen a marked rise in clinical trials of metronomic chemotherapy, and it is increasingly combined in the clinic with conventional treatments, such as maximum-tolerated dose chemotherapy and radiotherapy, as well as with novel therapeutic strategies, such as drug repositioning, targeted agents and immunotherapy. We review the latest advances in understanding the complex mechanisms of action of metronomic chemotherapy, and the recently identified factors associated with disease resistance. We comprehensively discuss the latest clinical data obtained from studies performed in both adult and paediatric populations, and highlight ongoing clinical trials. In this Review, we foresee the future developments of metronomic chemotherapy and specifically its potential role in the era of personalized medicine.
Collapse
Affiliation(s)
- Nicolas André
- Service d'Hématologie & Oncologie Pédiatrique, AP-HM, 264 rue Saint Pierre, 13385 Marseille, France
| | - Manon Carré
- INSERM UMR 911, Centre de Recherche en Oncologie Biologique et Oncopharmacologie, Aix-Marseille University, 27 Boulevard Jean Moulin, 13005 Marseille, France
| | - Eddy Pasquier
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, PO Box 81, Randwick NSW 2031, Australia
| |
Collapse
|
33
|
Lin C, Song W, Bi X, Zhao J, Huang Z, Li Z, Zhou J, Cai J, Zhao H. Recent advances in the ARID family: focusing on roles in human cancer. Onco Targets Ther 2014; 7:315-24. [PMID: 24570593 PMCID: PMC3933769 DOI: 10.2147/ott.s57023] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The human AT-rich interaction domain (ARID) family contains seven subfamilies and 15 members characterized by having an ARID. Members of the ARID family have the ability to regulate transcription and are involved in cell differentiation and proliferation. Accumulating evidence suggests that ARID family members are involved in cancer-related signaling pathways, highly mutated or differentially expressed in tumor tissues, and act as predictive factors for cancer prognosis or therapeutic outcome. Here we review the molecular biology and clinical studies concerned with the role played by the ARID family in cancer. This may contribute to our understanding of the initiation and progression of cancer from a novel point of view, as well as providing potential targets for cancer therapy.
Collapse
Affiliation(s)
- Chen Lin
- Department of Abdominal Surgical Oncology, Cancer Hospital, Beijing, People's Republic of China
| | - Wei Song
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xinyu Bi
- Department of Abdominal Surgical Oncology, Cancer Hospital, Beijing, People's Republic of China
| | - Jianjun Zhao
- Department of Abdominal Surgical Oncology, Cancer Hospital, Beijing, People's Republic of China
| | - Zhen Huang
- Department of Abdominal Surgical Oncology, Cancer Hospital, Beijing, People's Republic of China
| | - Zhiyu Li
- Department of Abdominal Surgical Oncology, Cancer Hospital, Beijing, People's Republic of China
| | - Jianguo Zhou
- Department of Abdominal Surgical Oncology, Cancer Hospital, Beijing, People's Republic of China
| | - Jianqiang Cai
- Department of Abdominal Surgical Oncology, Cancer Hospital, Beijing, People's Republic of China
| | - Hong Zhao
- Department of Abdominal Surgical Oncology, Cancer Hospital, Beijing, People's Republic of China
| |
Collapse
|
34
|
Li H, Yu G, Shi R, Lang B, Chen X, Xia D, Xiao H, Guo X, Guan W, Ye Z, Xiao W, Xu H. Cisplatin-induced epigenetic activation of miR-34a sensitizes bladder cancer cells to chemotherapy. Mol Cancer 2014; 13:8. [PMID: 24423412 PMCID: PMC4022035 DOI: 10.1186/1476-4598-13-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 01/04/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Accumulating evidence suggests a tumor suppressive role for miR-34a in human carcinogenesis. However, its precise biological role remains largely elusive. This study aimed to reveal the association of the miR-34a expression and its modulation of sensitivity to cisplatin in muscle-invasive bladder cancer (MIBC). METHODS miR-34a expression in MIBC cell lines and patient tissues was investigated using qPCR. The methylation analysis of miR-34a promoter region was performed by MassARRAY. Synthetic short single or double stranded RNA oligonucleotides and lentiviral vector were used to regulate miR-34a expression in MIBC cells to investigate its function in vitro and in vivo. RESULTS miR-34a expression was frequently decreased in MIBC tissues and cell lines through promoter hypermethylation while it was epigenetically increased in MIBC cells following cisplatin treatment. Increased miR-34a expression significantly sensitized MIBC cells to cisplatin and inhibited the tumorigenicity and proliferation of cancer cells in vitro and in vivo. Furthermore, we identified CD44 as being targeted by miR-34a in MIBC cells following cisplatin treatment, and increased CD44 expression could efficiently reverse the effect of miR-34a on MIBC cell proliferation, colongenic potential and chemosensitivity. CONCLUSIONS Cisplatin-based chemotherapy induced demethylation of miR-34a promoter and increased miR-34a expression, which in turn sensitized MIBC cells to cisplatin and decreased the tumorigenicity and proliferation of cancer cells that by reducing the production of CD44.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Wei Xiao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | | |
Collapse
|
35
|
Koshkin V, Yang BB, Krylov SN. Kinetics of MDR transport in tumor-initiating cells. PLoS One 2013; 8:e79222. [PMID: 24223908 PMCID: PMC3815210 DOI: 10.1371/journal.pone.0079222] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 09/25/2013] [Indexed: 12/31/2022] Open
Abstract
Multidrug resistance (MDR) driven by ABC (ATP binding cassette) membrane transporters is one of the major causes of treatment failure in human malignancy. MDR capacity is thought to be unevenly distributed among tumor cells, with higher capacity residing in tumor-initiating cells (TIC) (though opposite finding are occasionally reported). Functional evidence for enhanced MDR of TICs was previously provided using a "side population" assay. This assay estimates MDR capacity by a single parameter - cell's ability to retain fluorescent MDR substrate, so that cells with high MDR capacity ("side population") demonstrate low substrate retention. In the present work MDR in TICs was investigated in greater detail using a kinetic approach, which monitors MDR efflux from single cells. Analysis of kinetic traces obtained allowed for the estimation of both the velocity (V max) and affinity (K M) of MDR transport in single cells. In this way it was shown that activation of MDR in TICs occurs in two ways: through the increase of V max in one fraction of cells, and through decrease of K M in another fraction. In addition, kinetic data showed that heterogeneity of MDR parameters in TICs significantly exceeds that of bulk cells. Potential consequences of these findings for chemotherapy are discussed.
Collapse
Affiliation(s)
- Vasilij Koshkin
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario, Canada
| | - Burton B. Yang
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Sergey N. Krylov
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario, Canada
| |
Collapse
|
36
|
Lee HC, Ling QD, Yu WC, Hung CM, Kao TC, Huang YW, Higuchi A. Drug-resistant colon cancer cells produce high carcinoembryonic antigen and might not be cancer-initiating cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:491-502. [PMID: 23818760 PMCID: PMC3693723 DOI: 10.2147/dddt.s45890] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Purpose We evaluated the higher levels of carcinoembryonic antigen (CEA) secreted by the LoVo human colon carcinoma cells in a medium containing anticancer drugs. Drug-resistant LoVo cells were analyzed by subcutaneously xenotransplanting them into mice. The aim of this study was to evaluate whether the drug-resistant cells isolated in this study were cancer-initiating cells, known also as cancer stem cells (CSCs). Methods The production of CEA was investigated in LoVo cells that were cultured with 0–10 mM of anticancer drugs, and we evaluated the increase in CEA production by the LoVo cells that were stimulated by anticancer drug treatment. The expression of several CSC markers in LoVo cells treated with anticancer drugs was also evaluated. Following anticancer drug treatment, LoVo cells were injected subcutaneously into the flanks of severe combined immunodeficiency mice in order to evaluate the CSC fraction. Results Production of CEA by LoVo cells was stimulated by the addition of anticancer drugs. Drug-resistant LoVo cells expressed lower levels of CSC markers, and LoVo cells treated with any of the anticancer drugs tested did not generate tumors within 8 weeks from when the cells were injected subcutaneously into severe combined immunodeficiency mice. These results suggest that the drug-resistant LoVo cells have a smaller population of CSCs than the untreated LoVo cells. Conclusion Production of CEA by LoVo cells can be stimulated by the addition of anticancer drugs. The drug-resistant subpopulation of LoVo colon cancer cells could stimulate the production of CEA, but these cells did not act as CSCs in in vivo tumor generation experiments.
Collapse
Affiliation(s)
- Hsin-chung Lee
- Graduate Institute of Systems Biology and Bioinformatics, National Central University, Jhongli, Taoyuan
| | | | | | | | | | | | | |
Collapse
|
37
|
Yang DR, Ding XF, Luo J, Shan YX, Wang R, Lin SJ, Li G, Huang CK, Zhu J, Chen Y, Lee SO, Chang C. Increased chemosensitivity via targeting testicular nuclear receptor 4 (TR4)-Oct4-interleukin 1 receptor antagonist (IL1Ra) axis in prostate cancer CD133+ stem/progenitor cells to battle prostate cancer. J Biol Chem 2013; 288:16476-16483. [PMID: 23609451 DOI: 10.1074/jbc.m112.448142] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer (PCa) stem/progenitor cells are known to have higher chemoresistance than non-stem/progenitor cells, but the underlying molecular mechanism remains unclear. We found the expression of testicular nuclear receptor 4 (TR4) is significantly higher in PCa CD133(+) stem/progenitor cells compared with CD133(-) non-stem/progenitor cells. Knockdown of TR4 levels in the established PCa stem/progenitor cells and the CD133(+) population of the C4-2 PCa cell line with lentiviral TR4 siRNA led to increased drug sensitivity to the two commonly used chemotherapeutic drugs, docetaxel and etoposide, judging from significantly reduced IC50 values and increased apoptosis in the TR4 knockdown cells. Mechanism dissection studies found that suppression of TR4 in these stem/progenitor cells led to down-regulation of Oct4 expression, which, in turn, down-regulated the IL-1 receptor antagonist (IL1Ra) expression. Neutralization experiments via adding these molecules into the TR4 knockdown PCa stem/progenitor cells reversed the chemoresistance, suggesting that the TR4-Oct4-IL1Ra axis may play a critical role in the development of chemoresistance in the PCa stem/progenitor cells. Together, these studies suggest that targeting TR4 may alter chemoresistance of PCa stem/progenitor cells, and this finding provides the possibility of targeting TR4 as a new and better approach to overcome the chemoresistance problem in PCa therapeutics.
Collapse
Affiliation(s)
- Dong-Rong Yang
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and the Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14642; Department of Urology, Second Affiliated Hospital of Soochow University, Suzhou, 215004 China
| | - Xian-Fan Ding
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and the Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14642; Department of Urology, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, 310016 China
| | - Jie Luo
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and the Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14642
| | - Yu-Xi Shan
- Department of Urology, Second Affiliated Hospital of Soochow University, Suzhou, 215004 China
| | - Ronghao Wang
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and the Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14642
| | - Shin-Jen Lin
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and the Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14642
| | - Gonghui Li
- Department of Urology, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, 310016 China
| | - Chiung-Kuei Huang
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and the Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14642
| | - Jin Zhu
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and the Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14642; Department of Urology, Second Affiliated Hospital of Soochow University, Suzhou, 215004 China
| | - Yuhchyau Chen
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and the Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14642
| | - Soo Ok Lee
- Department of Urology, Second Affiliated Hospital of Soochow University, Suzhou, 215004 China.
| | - Chawnshang Chang
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology, and Radiation Oncology and the Wilmot Cancer Center. University of Rochester Medical Center, Rochester, New York 14642; Sex Hormone Research Center, China Medical University/Hospital, Taichung 404, Taiwan.
| |
Collapse
|
38
|
Koshkin V, Krylov SN. Single-Cell-Kinetics Approach to Discover Functionally Distinct Subpopulations within Phenotypically Uniform Populations of Cells. Anal Chem 2013; 85:2578-81. [DOI: 10.1021/ac400151v] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Vasilij Koshkin
- Department of Chemistry and Centre
for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| | - Sergey N. Krylov
- Department of Chemistry and Centre
for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| |
Collapse
|
39
|
|
40
|
Hegde GV, de la Cruz C, Eastham-Anderson J, Zheng Y, Sweet-Cordero EA, Jackson EL. Residual tumor cells that drive disease relapse after chemotherapy do not have enhanced tumor initiating capacity. PLoS One 2012; 7:e45647. [PMID: 23115623 PMCID: PMC3480356 DOI: 10.1371/journal.pone.0045647] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 08/20/2012] [Indexed: 12/17/2022] Open
Abstract
Although chemotherapy is used to treat most advanced solid tumors, recurrent disease is still the major cause of cancer-related mortality. Cancer stem cells (CSCs) have been the focus of intense research in recent years because they provide a possible explanation for disease relapse. However, the precise role of CSCs in recurrent disease remains poorly understood and surprisingly little attention has been focused on studying the cells responsible for re-initiating tumor growth within the original host after chemotherapy treatment. We utilized both xenograft and genetically engineered mouse models of non-small cell lung cancer (NSCLC) to characterize the residual tumor cells that survive chemotherapy treatment and go on to cause tumor regrowth, which we refer to as tumor re-initiating cells (TRICs). We set out to determine whether TRICs display characteristics of CSCs, and whether assays used to define CSCs also provide an accurate readout of a cell's ability to cause tumor recurrence. We did not find consistent enrichment of CSC marker positive cells or enhanced tumor initiating potential in TRICs. However, TRICs from all models do appear to be in EMT, a state that has been linked to chemoresistance in numerous types of cancer. Thus, the standard CSC assays may not accurately reflect a cell's ability to drive disease recurrence.
Collapse
Affiliation(s)
- Ganapati V. Hegde
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - Cecile de la Cruz
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - Jeffrey Eastham-Anderson
- Department of Pathology, Genentech, Inc., South San Francisco, California, United States of America
| | - Yanyan Zheng
- Cancer Biology Program, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, United States of America
| | - E. Alejandro Sweet-Cordero
- Cancer Biology Program, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Erica L. Jackson
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
Abstract
Heterogeneity is an omnipresent feature of mammalian cells in vitro and in vivo. It has been recently realized that even mouse and human embryonic stem cells under the best culture conditions are heterogeneous containing pluripotent as well as partially committed cells. Somatic stem cells in adult organs are also heterogeneous, containing many subpopulations of self-renewing cells with distinct regenerative capacity. The differentiated progeny of adult stem cells also retain significant developmental plasticity that can be induced by a wide variety of experimental approaches. Like normal stem cells, recent data suggest that cancer stem cells (CSCs) similarly display significant phenotypic and functional heterogeneity, and that the CSC progeny can manifest diverse plasticity. Here, I discuss CSC heterogeneity and plasticity in the context of tumor development and progression, and by comparing with normal stem cell development. Appreciation of cancer cell plasticity entails a revision to the earlier concept that only the tumorigenic subset in the tumor needs to be targeted. By understanding the interrelationship between CSCs and their differentiated progeny, we can hope to develop better therapeutic regimens that can prevent the emergence of tumor cell variants that are able to found a new tumor and distant metastases.
Collapse
|