51
|
Debnath S, Debnath T, Bhaumik S, Majumdar S, Kalle AM, Aparna V. Discovery of novel potential selective HDAC8 inhibitors by combine ligand-based, structure-based virtual screening and in-vitro biological evaluation. Sci Rep 2019; 9:17174. [PMID: 31748509 PMCID: PMC6868012 DOI: 10.1038/s41598-019-53376-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 10/31/2019] [Indexed: 12/31/2022] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor found in children and survival rate is extremely meager. HDAC8, a class I zinc-dependent enzyme, is a potential drug target for treatment of neuroblastoma and T cell lymphoma. Most of the HDAC8 inhibitors discovered till date contains a hydroxamic acid group which acts as a zinc binding group. The high binding affinity to the zinc and other ions results in adverse effects. Also, the non-selective inhibition of HDACs cause a variety of side effects. The objective of this is to identify structurally diverse, non-hydroxamate, novel, potential and selective HDAC8 inhibitors. A number of five featured pharmacophore hypotheses were generated using 32 known selective HDAC8 inhibitors. The hypotheses ADDRR.4 were selected for building 3D QSAR model. This model has an excellent correlation coefficient and good predictive ability, which was employed for virtual screening of Phase database containing 4.3 × 106 molecules. The resultant hits with fitness score >1.0 were optimized using in-silico ADMET (absorption, distribution, metabolism, excretion, and toxicity) and XP glide docking studies. On the basis of pharmacophore matching, interacting amino acid residues, XP glide score, more affinity towards HDAC8 and less affinity towards other HDACs, and ADME results five hits- SD-01, SD-02, SD-03, SD-04 and SD-05 with new structural scaffolds, non-hydroxamate were selected for in vitro activity study. SD-01 and SD-02 were found to be active in the nanomolar (nM) range. SD-01 had considerably good selectivity for HDAC8 over HDAC6 and SD-02 had marginal selectivity for HDAC6 over HDAC8. The compounds SD-01 and SD-02 were found to inhibit HDAC8 at concentrations (IC50) 9.0 nM and 2.7 nM, respectively.
Collapse
Affiliation(s)
- Sudhan Debnath
- Department of Chemistry, MBB College, Agartala, Tripura, 799004, India.
| | - Tanusree Debnath
- Department of Chemistry, MBB College, Agartala, Tripura, 799004, India
| | - Samhita Bhaumik
- Department of Chemistry, Women's College, Agartala, Tripura, 799001, India
| | - Swapan Majumdar
- Department of Chemistry, Tripura University, Suryamaninagar, Agartala, Tripura, 799022, India
| | - Arunasree M Kalle
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, TS, 500046, India
| | - Vema Aparna
- Sree Chaitanya Institute of Pharmaceutical Sciences, Karimnagar, 505 527, Andhra Pradesh, India
| |
Collapse
|
52
|
Peng X, Liao G, Sun P, Yu Z, Chen J. An Overview of HDAC Inhibitors and their Synthetic Routes. Curr Top Med Chem 2019; 19:1005-1040. [DOI: 10.2174/1568026619666190227221507] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/19/2019] [Accepted: 01/28/2019] [Indexed: 12/21/2022]
Abstract
Epigenetics play a key role in the origin, development and metastasis of cancer. Epigenetic processes include DNA methylation, histone acetylation, histone methylation, and histone phosphorylation, among which, histone acetylation is the most common one that plays important roles in the regulation of normal cellular processes, and is controlled by histone deacetylases (HDACs) and histone acetyltransferases (HATs). HDACs are involved in the regulation of many key cellular processes, such as DNA damage repair, cell cycle control, autophagy, metabolism, senescence and chaperone function, and can lead to oncogene activation. As a result, HDACs are considered to be an excellent target for anti-cancer therapeutics like histone deacetylase inhibitors (HDACi) which have attracted much attention in the last decade. A wide-ranging knowledge of the role of HDACs in tumorigenesis, and of the action of HDACi, has been achieved. The primary purpose of this paper is to summarize recent HDAC inhibitors and the synthetic routes as well as to discuss the direction for the future development of new HDAC inhibitors.
Collapse
Affiliation(s)
- Xiaopeng Peng
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Guochao Liao
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Pinghua Sun
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
53
|
Vivet-Noguer R, Tarin M, Roman-Roman S, Alsafadi S. Emerging Therapeutic Opportunities Based on Current Knowledge of Uveal Melanoma Biology. Cancers (Basel) 2019; 11:E1019. [PMID: 31330784 PMCID: PMC6678734 DOI: 10.3390/cancers11071019] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/09/2019] [Accepted: 07/17/2019] [Indexed: 12/16/2022] Open
Abstract
Uveal Melanoma (UM) is a rare and malignant intraocular tumor with dismal prognosis. Despite the efficient control of the primary tumor by radiation or surgery, up to 50% of patients subsequently develop metastasis, mainly in the liver. Once the tumor has spread from the eye, the treatment is challenging and the median survival is only nine months. UM represents an intriguing model of oncogenesis that is characterized by a relatively homogeneous histopathological architecture and a low burden of genetic alterations, in contrast to other melanomas. UM is driven by recurrent activating mutations in Gαq pathway, which are associated with a second mutation in BRCA1 associated protein 1 (BAP1), splicing factor 3b subunit 1 (SF3B1), or eukaryotic translation initiation factor 1A X-linked (EIF1AX), occurring in an almost mutually exclusive manner. The monosomy of chromosome 3 is also a recurrent feature that is associated with high metastatic risk. These events driving UM oncogenesis have been thoroughly investigated over the last decade. However, no efficient related therapeutic strategies are yet available and the metastatic disease remains mostly incurable. Here, we review current knowledge regarding the molecular biology and the genetics of uveal melanoma and highlight the related therapeutic applications and perspectives.
Collapse
Affiliation(s)
- Raquel Vivet-Noguer
- Uveal Melanoma Translational Group, Department of Translational Research, Institut Curie, PSL Research University, 75248 Paris, France
| | - Malcy Tarin
- Uveal Melanoma Translational Group, Department of Translational Research, Institut Curie, PSL Research University, 75248 Paris, France
| | - Sergio Roman-Roman
- Uveal Melanoma Translational Group, Department of Translational Research, Institut Curie, PSL Research University, 75248 Paris, France
| | - Samar Alsafadi
- Uveal Melanoma Translational Group, Department of Translational Research, Institut Curie, PSL Research University, 75248 Paris, France.
| |
Collapse
|
54
|
p53 at the Crossroads between Different Types of HDAC Inhibitor-Mediated Cancer Cell Death. Int J Mol Sci 2019; 20:ijms20102415. [PMID: 31096697 PMCID: PMC6567317 DOI: 10.3390/ijms20102415] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022] Open
Abstract
Cancer is a complex genetic and epigenetic-based disease that has developed an armada of mechanisms to escape cell death. The deregulation of apoptosis and autophagy, which are basic processes essential for normal cellular activity, are commonly encountered during the development of human tumors. In order to assist the cancer cell in defeating the imbalance between cell growth and cell death, histone deacetylase inhibitors (HDACi) have been employed to reverse epigenetically deregulated gene expression caused by aberrant post-translational protein modifications. These interfere with histone acetyltransferase- and deacetylase-mediated acetylation of both histone and non-histone proteins, and thereby exert a wide array of HDACi-stimulated cytotoxic effects. Key determinants of HDACi lethality that interfere with cellular growth in a multitude of tumor cells are apoptosis and autophagy, which are either mutually exclusive or activated in combination. Here, we compile known molecular signals and pathways involved in the HDACi-triggered induction of apoptosis and autophagy. Currently, the factors that determine the mode of HDACi-elicited cell death are mostly unclear. Correspondingly, we also summarized as yet established intertwined mechanisms, in particular with respect to the oncogenic tumor suppressor protein p53, that drive the interplay between apoptosis and autophagy in response to HDACi. In this context, we also note the significance to determine the presence of functional p53 protein levels in the cancer cell. The confirmation of the context-dependent function of autophagy will pave the way to improve the benefit from HDACi-mediated cancer treatment.
Collapse
|
55
|
YADAV R, MISHRA P, YADAV D. Histone Deacetylase Inhibitors: A Prospect in Drug Discovery. Turk J Pharm Sci 2019; 16:101-114. [PMID: 32454703 PMCID: PMC7227979 DOI: 10.4274/tjps.75047] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
Abstract
Cancer is a provocative issue across the globe and treatment of uncontrolled cell growth follows a deep investigation in the field of drug discovery. Therefore, there is a crucial requirement for discovering an ingenious medicinally active agent that can amend idle drug targets. Increasing pragmatic evidence implies that histone deacetylases (HDACs) are trapped during cancer progression, which increases deacetylation and triggers changes in malignancy. They provide a ground-breaking scaffold and an attainable key for investigating chemical entity pertinent to HDAC biology as a therapeutic target in the drug discovery context. Due to gene expression, an impending requirement to prudently transfer cytotoxicity to cancerous cells, HDAC inhibitors may be developed as anticancer agents. The present review focuses on the basics of HDAC enzymes, their inhibitors, and therapeutic outcomes.
Collapse
Affiliation(s)
- Rakesh YADAV
- Banasthali University, Faculty of Pharmacy, Department of Pharmacy, Banasthali, India
| | - Pooja MISHRA
- Banasthali University, Faculty of Pharmacy, Department of Pharmacy, Banasthali, India
| | - Divya YADAV
- Banasthali University, Faculty of Pharmacy, Department of Pharmacy, Banasthali, India
| |
Collapse
|
56
|
Zhang Q, Wang S, Chen J, Yu Z. Histone Deacetylases (HDACs) Guided Novel Therapies for T-cell lymphomas. Int J Med Sci 2019; 16:424-442. [PMID: 30911277 PMCID: PMC6428980 DOI: 10.7150/ijms.30154] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022] Open
Abstract
T-cell lymphomas are a heterogeneous group of cancers with different pathogenesis and poor prognosis. Histone deacetylases (HDACs) are epigenetic modifiers that modulate many key biological processes. In recent years, HDACs have been fully investigated for their roles and potential as drug targets in T-cell lymphomas. In this review, we have deciphered the modes of action of HDACs, HDAC inhibitors as single agents, and HDACs guided combination therapies in T-cell lymphomas. The overview of HDACs on the stage of T-cell lymphomas, and HDACs guided therapies both as single agents and combination regimens endow great opportunities for the cure of T-cell lymphomas.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Shaobin Wang
- Health Management Center of Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Junhui Chen
- Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Zhendong Yu
- China Central Laboratory of Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| |
Collapse
|
57
|
Epigenetic Targeting of Autophagy via HDAC Inhibition in Tumor Cells: Role of p53. Int J Mol Sci 2018; 19:ijms19123952. [PMID: 30544838 PMCID: PMC6321134 DOI: 10.3390/ijms19123952] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 12/19/2022] Open
Abstract
Tumor development and progression is the consequence of genetic as well as epigenetic alterations of the cell. As part of the epigenetic regulatory system, histone acetyltransferases (HATs) and deacetylases (HDACs) drive the modification of histone as well as non-histone proteins. Derailed acetylation-mediated gene expression in cancer due to a delicate imbalance in HDAC expression can be reversed by histone deacetylase inhibitors (HDACi). Histone deacetylase inhibitors have far-reaching anticancer activities that include the induction of cell cycle arrest, the inhibition of angiogenesis, immunomodulatory responses, the inhibition of stress responses, increased generation of oxidative stress, activation of apoptosis, autophagy eliciting cell death, and even the regulation of non-coding RNA expression in malignant tumor cells. However, it remains an ongoing issue how tumor cells determine to respond to HDACi treatment by preferentially undergoing apoptosis or autophagy. In this review, we summarize HDACi-mediated mechanisms of action, particularly with respect to the induction of cell death. There is a keen interest in assessing suitable molecular factors allowing a prognosis of HDACi-mediated treatment. Addressing the results of our recent study, we highlight the role of p53 as a molecular switch driving HDACi-mediated cellular responses towards one of both types of cell death. These findings underline the importance to determine the mutational status of p53 for an effective outcome in HDACi-mediated tumor therapy.
Collapse
|
58
|
Raghunath A, Sundarraj K, Arfuso F, Sethi G, Perumal E. Dysregulation of Nrf2 in Hepatocellular Carcinoma: Role in Cancer Progression and Chemoresistance. Cancers (Basel) 2018; 10:cancers10120481. [PMID: 30513925 PMCID: PMC6315366 DOI: 10.3390/cancers10120481] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/29/2018] [Accepted: 11/29/2018] [Indexed: 12/23/2022] Open
Abstract
The liver executes versatile functions and is the chief organ for metabolism of toxicants/xenobiotics. Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and the third foremost cause of cancer death worldwide. Oxidative stress is a key factor related with the development and progression of HCC. Nuclear factor erythroid 2 [NF-E2]-related factor 2 (Nrf2) is a cytosolic transcription factor, which regulates redox homeostasis by activating the expression of an array of antioxidant response element-dependent genes. Nrf2 displays conflicting roles in normal, healthy liver and HCC; in the former, Nrf2 offers beneficial effects, whereas in the latter it causes detrimental effects favouring the proliferation and survival of HCC. Sustained Nrf2 activation has been observed in HCC and facilitates its progression and aggressiveness. This review summarizes the role and mechanism(s) of action of Nrf2 dysregulation in HCC and therapeutic options that can be employed to modulate this transcription factor.
Collapse
Affiliation(s)
- Azhwar Raghunath
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, Tamilnadu, India.
| | - Kiruthika Sundarraj
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, Tamilnadu, India.
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6009, Australia.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, Tamilnadu, India.
| |
Collapse
|
59
|
Zarkesh M, Zadeh-Vakili A, Azizi F, Foroughi F, Akhavan MM, Hedayati M. Altered Epigenetic Mechanisms in Thyroid Cancer Subtypes. Mol Diagn Ther 2018; 22:41-56. [PMID: 28986854 DOI: 10.1007/s40291-017-0303-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Thyroid carcinoma (TC) is the most frequent malignant neoplasm of the endocrine system. Molecular methods for diagnosis of invasive thyroid disease can be effectively adopted. Epigenetic factors play an important role in the diversity patterns of gene expression and the phenotypic and biological characteristics of TC subtypes. We aimed to review epigenetic changes in the main subtypes of TC, along with a presentation of the methods that have examined these changes, and active clinical trials for the treatment of advanced TCs targeting epigenetic changes. A literature analysis was performed in MEDLINE using PubMed, Elsevier, and Google Scholar for studies published up to 2016, using the keywords: "Epigenetic alterations" OR "Epigenetic changes", "thyroid cancers", "papillary thyroid cancer", "medullary thyroid cancer", "follicular thyroid cancer", and "anaplastic thyroid cancer", which resulted in 310 articles in English. All related abstracts were reviewed and studies were included that were published in English, had available full text, and determined the details of the methods and materials associated with the epigenetic patterns of TC and its subtypes (100 articles). Analysis of epigenetic alterations in TC subtypes helps to identify pathogenesis and can play an important role in the classification and diagnosis of tumors. Epigenetic mechanisms, especially aberrant methylation of DNA and microRNAs (miRs), are likely to play an important role in thyroid tumorigenesis. Further studies are required to elucidate the role of histone modification mechanisms in TC development.
Collapse
Affiliation(s)
- Maryam Zarkesh
- Cellular and Molecular Endocrine Research Center (CMERC), Research Institute for Endocrine Sciences of Shahid Beheshti University of Medical Sciences, 19395-4763, Tehran, Iran
| | - Azita Zadeh-Vakili
- Cellular and Molecular Endocrine Research Center (CMERC), Research Institute for Endocrine Sciences of Shahid Beheshti University of Medical Sciences, 19395-4763, Tehran, Iran.
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Forough Foroughi
- Department of Pathology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maziar Mohammad Akhavan
- Skin Research Center School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center (CMERC), Research Institute for Endocrine Sciences of Shahid Beheshti University of Medical Sciences, 19395-4763, Tehran, Iran.
| |
Collapse
|
60
|
Wang X, Shen C, Liu Z, Peng F, Chen X, Yang G, Zhang D, Yin Z, Ma J, Zheng Z, Zhao B, Liu H, Wang L, Wu J, Han D, Wang K, Zhong C, Hou X, Zhao W, Shu M, Wang X, Zhao S. Nitazoxanide, an antiprotozoal drug, inhibits late-stage autophagy and promotes ING1-induced cell cycle arrest in glioblastoma. Cell Death Dis 2018; 9:1032. [PMID: 30302016 PMCID: PMC6177448 DOI: 10.1038/s41419-018-1058-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 08/07/2018] [Accepted: 09/03/2018] [Indexed: 11/09/2022]
Abstract
Glioblastoma is the most common and aggressive primary brain tumor in adults. New drug design and development is still a major challenge for glioma treatment. Increasing evidence has shown that nitazoxanide, an antiprotozoal drug, has a novel antitumor role in various tumors and exhibits multiple molecular functions, especially autophagic regulation. However, whether nitazoxanide-associated autophagy has an antineoplastic effect in glioma remains unclear. Here, we aimed to explore the underlying molecular mechanism of nitazoxanide in glioblastoma. Our results showed that nitazoxanide suppressed cell growth and induced cell cycle arrest in glioblastoma by upregulating ING1 expression with a favorable toxicity profile. Nitazoxanide inhibited autophagy through blockage of late-stage lysosome acidification, resulting in decreased cleavage of ING1. A combination with chloroquine or Torin1 enhanced or impaired the chemotherapeutic effect of nitazoxanide in glioblastoma cells. Taken together, these findings indicate that nitazoxanide as an autophagy inhibitor induces cell cycle arrest in glioblastoma via upregulated ING1 due to increased transcription and decreased post-translational degradation by late-stage autophagic inhibition.
Collapse
Affiliation(s)
- Xiaoxiong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Chen Shen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Zhendong Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Fei Peng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Guang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Daming Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Zhiqin Yin
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Jichao Ma
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, No. 157 Baojian Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Zhixing Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Boxian Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Huailei Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Ligang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Jianing Wu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Dayong Han
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Kaikai Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Chen Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Xu Hou
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Wenyang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Mengting Shu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Xinzhuang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Shiguang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China. .,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China. .,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
61
|
Li T, Zhang C, Hassan S, Liu X, Song F, Chen K, Zhang W, Yang J. Histone deacetylase 6 in cancer. J Hematol Oncol 2018; 11:111. [PMID: 30176876 PMCID: PMC6122547 DOI: 10.1186/s13045-018-0654-9] [Citation(s) in RCA: 221] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 08/22/2018] [Indexed: 12/15/2022] Open
Abstract
Histone acetylation and deacetylation are important epigenetic mechanisms that regulate gene expression and transcription. Histone deacetylase 6 (HDAC6) is a unique member of the HDAC family that not only participates in histone acetylation and deacetylation but also targets several nonhistone substrates, such as α-tubulin, cortactin, and heat shock protein 90 (HSP90), to regulate cell proliferation, metastasis, invasion, and mitosis in tumors. Furthermore, HDAC6 also upregulates several critical factors in the immune system, such as program death receptor-1 (PD-1) and program death receptor ligand-1 (PD-L1) receptor, which are the main targets for cancer immunotherapy. Several selective HDAC6 inhibitors are currently in clinical trials for cancer treatment and bring hope for patients with malignant tumors. A fuller understanding of HDAC6 as a critical regulator of many cellular pathways will help further the development of targeted anti-HDAC6 therapies. Here, we review the unique features of HDAC6 and its role in cancer, which make HDAC6 an appealing drug target.
Collapse
Affiliation(s)
- Ting Li
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China
| | - Shafat Hassan
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.,International Medical School, Tianjin Medical University, Tianjin, 300061, People's Republic of China
| | - Xinyue Liu
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China
| | - Fengju Song
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China
| | - Wei Zhang
- Cancer Genomics and Precision Medicine, Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Jilong Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China. .,National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China.
| |
Collapse
|
62
|
Schiaffino L, Bonafede R, Scambi I, Parrella E, Pizzi M, Mariotti R. Acetylation state of RelA modulated by epigenetic drugs prolongs survival and induces a neuroprotective effect on ALS murine model. Sci Rep 2018; 8:12875. [PMID: 30150770 PMCID: PMC6110772 DOI: 10.1038/s41598-018-30659-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
Dysregulation in acetylation homeostasis has been implicated in the pathogenesis of the amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disorder. It is known that the acetylation of transcriptional factors regulates their activity. The acetylation state of NF-kB RelA has been found to dictate the neuroprotective versus the neurotoxic effect of p50/RelA. Here we showed that the pro-apoptotic acetylation mode of RelA, involving a general lysine deacetylation of the subunit with the exclusion of the lysine 310, is evident in the lumbar spinal cord of SOD1(G93A) mice, a murine model of ALS. The administration of the HDAC inhibitor MS-275 and the AMPK/sirtuin 1 activator resveratrol restored the normal RelA acetylation in SOD1(G93A) mice. The SOD1(G93A) mice displayed a 3 weeks delay of the disease onset, associated with improvement of motor performance, and 2 weeks increase of lifespan. The epigenetic treatment rescued the lumbar motor neurons affected in SOD1(G93A) mice, accompanied by increased levels of protein products of NF-kB-target genes, Bcl-xL and brain-derived neurotrophic factor. In conclusion, we here demonstrate that MS-275 and resveratrol restore the acetylation state of RelA in the spinal cord, delaying the onset and increasing the lifespan of SOD1(G93A) mice.
Collapse
Affiliation(s)
- Lorenzo Schiaffino
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Roberta Bonafede
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Ilaria Scambi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Edoardo Parrella
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Raffaella Mariotti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| |
Collapse
|
63
|
He S, Dong G, Wu S, Fang K, Miao Z, Wang W, Sheng C. Small Molecules Simultaneously Inhibiting p53-Murine Double Minute 2 (MDM2) Interaction and Histone Deacetylases (HDACs): Discovery of Novel Multitargeting Antitumor Agents. J Med Chem 2018; 61:7245-7260. [PMID: 30045621 DOI: 10.1021/acs.jmedchem.8b00664] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
p53-Murine double minute 2 (MDM2) interaction and histone deacetylases (HDACs) are important targets in antitumor drug development. Inspired by the synergistic effects between MDM2 and HDACs, the first MDM2/HDACs dual inhibitors were identified, which showed excellent activities against both targets. In particular, compound 14d was proven to be a potent and orally active MDM2/HDAC dual inhibitor, whose antitumor mechanisms were validated in cancer cells. Compound 14d showed excellent in vivo antitumor potency in the A549 xenograft model, providing a promising lead compound for the development of novel antitumor agents. Also, this proof-of-concept study offers a novel and efficient strategy for multitargeting antitumor drug discovery.
Collapse
Affiliation(s)
- Shipeng He
- School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , P.R. China
| | - Guoqiang Dong
- Department of Medicinal Chemistry, School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , P.R. China
| | - Shanchao Wu
- Department of Medicinal Chemistry, School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , P.R. China
| | - Kun Fang
- School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , P.R. China
| | - Zhenyuan Miao
- Department of Medicinal Chemistry, School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , P.R. China
| | - Wei Wang
- School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , P.R. China.,Department of Chemistry and Chemical Biology , University of New Mexico , MSC03 2060, Albuquerque , New Mexico 87131-0001 , United States
| | - Chunquan Sheng
- Department of Medicinal Chemistry, School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , P.R. China
| |
Collapse
|
64
|
Wang B, Lyu H, Pei S, Song D, Ni J, Liu B. Cladribine in combination with entinostat synergistically elicits anti-proliferative/anti-survival effects on multiple myeloma cells. Cell Cycle 2018; 17:985-996. [PMID: 29969371 PMCID: PMC6197031 DOI: 10.1080/15384101.2018.1464849] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 04/08/2018] [Indexed: 12/28/2022] Open
Abstract
Cladribine (2CdA), a synthetic purine analog interfering with DNA synthesis, is a medication used to treat hairy cell leukemia (HCL) and B-cell chronic lymphocytic leukemia. Entinostat, a selective class I histone deacetylase (HDAC) inhibitor, shows antitumor activity in various human cancers, including hematological malignancies. The therapeutic potential of cladribine and entinostat against multiple myeloma (MM) remains unclear. Here we investigate the combinatorial effects of cladribine and entinostat within the range of their clinical achievable concentrations on MM cells. While either agent alone inhibited MM cell proliferation in a dose-dependent manner, their combinations synergistically induced anti-proliferative/anti-survival effects on all MM cell lines (RPMI8226, U266, and MM1.R) tested. Further studies showed that the combinations of cladribine and entinostat as compared to either agent alone more potently induced mitotic catastrophe in the MM cells, and resulted in a marked increase of the cells at G1 phase associated with decrease of Cyclin D1 and E2F-1 expression and upregulation of p21waf-1. Apoptotic ELISA and western blot analyses revealed that the combinations of cladribine and entinostat exerted a much more profound activity to induce apoptosis and DNA damage response, evidenced by enhanced phosphorylation of histone H2A.X and the DNA repair enzymes Chk1 and Chk2. Collectively, our data demonstrate that the combinations of cladribine and entinostat exhibit potent activity to induce anti-proliferative/anti-survival effects on MM cells via induction of cell cycle G1 arrest, apoptosis, and DNA damage response. Regimens consisting of cladribine and/or entinostat may offer a new treatment option for patients with MM. ABBREVIATIONS MM, multiple myeloma; HCL, hairy cell leukemia; HDAC, histone deacetylase; Ab, antibody; mAb, monoclonal Ab; FBS, fetal bovine serum; CI, combination index; PAGE, polyacrylamide gel electrophoresis; ELISA, enzyme-linked immunosorbent assay; PARP, poly(ADP-ribose) polymerase; MTS, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium,inner salt.
Collapse
Affiliation(s)
- Bolun Wang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hui Lyu
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Shanshan Pei
- Department of Hematology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Deye Song
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiangdong Ni
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bolin Liu
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
65
|
Barneh F, Salimi M, Goshadrou F, Ashtiani M, Mirzaie M, Zali H, Jafari M. Valproic acid inhibits the protective effects of stromal cells against chemotherapy in breast cancer: Insights from proteomics and systems biology. J Cell Biochem 2018; 119:9270-9283. [PMID: 29953653 DOI: 10.1002/jcb.27196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/24/2018] [Indexed: 12/14/2022]
Abstract
Interaction between tumor and stromal cells is beginning to be decoded as a contributor to chemotherapy resistance. Here, we aim to take a system-level approach to explore a mechanism by which stromal cells induce chemoresistance in cancer cells and subsequently identify a drug that can inhibit such interaction. Using a proteomic dataset containing quantitative data on secretome of stromal cells, we performed multivariate analyses and found that bone-marrow mesenchymal stem cells (BM-MSCs) play the most protective role against chemotherapeutics. Pathway enrichment tests showed that secreted cytokines from BM-MSCs activated 4 signaling pathways including Janus kinase-signal transducer and activator of transcription, phosphatidylinositol 3-kinase-protein kinase B, and mitogen-activated protein kinase, transforming growth factor-β in cancer cells collectively leading to nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) transcription factor activation. Based on the data from integrated Library of Integrated Network-Based Cellular Signatures (iLINCs) program, we found that among different drugs, valproic acid (VA) affected the expression of 34 genes within the identified pathways that are activated by stromal cells. Our in vitro experiments confirmed that VA inhibits NF-kB activation in cancer cells. In addition, analyzing gene expression data in patients taking oral VA showed that this drug decreased expression of antioxidant enzymes culminating in increased oxidative stress in tumor cells. These results suggest that VA confines the protective role of stromal cells by inhibiting the adaptation mechanisms toward oxidative stress which is potentiated by stromal cells. Since VA is an already prescribed drug manifesting anticancer effects, this study provides a mechanistic insight for combination of VA with chemotherapy in the clinical setting.
Collapse
Affiliation(s)
- Farnaz Barneh
- Department of Basic Sciences, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Physiology and Pharmacology Department, Pasteur Institute of Iran, Tehran, Iran.,Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mona Salimi
- Physiology and Pharmacology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Goshadrou
- Department of Basic Sciences, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Minoo Ashtiani
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.,Department of Computer Science and Statistics, Faculty of Mathematics, K. N. Toosi University of Technology, Tehran, Iran
| | - Mehdi Mirzaie
- Department of Applied Mathematics, Faculty of Mathematical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hakimeh Zali
- School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical sciences, Tehran, Iran
| | - Mohieddin Jafari
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
66
|
Jing B, Jin J, Xiang R, Liu M, Yang L, Tong Y, Xiao X, Lei H, Liu W, Xu H, Deng J, Zhou L, Wu Y. Vorinostat and quinacrine have synergistic effects in T-cell acute lymphoblastic leukemia through reactive oxygen species increase and mitophagy inhibition. Cell Death Dis 2018; 9:589. [PMID: 29789603 PMCID: PMC5964102 DOI: 10.1038/s41419-018-0679-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/05/2018] [Accepted: 05/07/2018] [Indexed: 12/16/2022]
Abstract
Despite recent progress in the treatment, the outcome of adult acute T-cell lymphoblastic leukemia (T-ALL) is poor. Development of novel approach to combat this disease is urgently required. Vorinostat, a pan-histone deacetylase (HDAC) inhibitor, exerts promising anticancer activity in a variety of solid and hematologic malignancies. However, the efficacy of vorinostat monotherapy is unsatisfactory. Here, we show that quinacrine (QC), an anti-malaria drug with potent autophagy inhibitory activity, could synergistically enhance vorinostat-induced cell death at a non-toxic concentration. Compared to the single treatment, QC plus vorinostat significantly induced apoptosis, disrupted the mitochondrial transmembrane potential, and decreased Mcl-1 and Bcl-2/Bax ratio. Interestingly, the application of QC plus vorinostat resulted in mitophagy blockade, as reflected by the increase in the K63-linked ubiquitination of mitochondria protein and the formation of mitochondrial aggresomes. QC plus vorinostat markedly increased the reactive oxygen species (ROS) level in cells. Moreover, the ROS scavenger N-acetylcysteine (NAC) abrogated QC plus vorinostat-induced ROS, decreased the ubiquitination of mitochondria proteins, and cell death. Finally, using a xenograft mouse model, we demonstrated that QC plus vorinostat significantly reduced cell proliferation and induced cell death in vivo. Taken together, our results showed that the combination of QC with vorinostat may represent a novel regimen for the treatment of T-cell acute lymphoblastic leukemia, which deserves clinical evaluation in the future.
Collapse
Affiliation(s)
- Bo Jing
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Chemical Biology Division of Shanghai Universities E-Institutes, Shanghai Tongren Hospital/Faculty of Basic Medicine, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jin Jin
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Chemical Biology Division of Shanghai Universities E-Institutes, Shanghai Tongren Hospital/Faculty of Basic Medicine, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Rufang Xiang
- Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin Er Road, Shanghai, China
| | - Meng Liu
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Chemical Biology Division of Shanghai Universities E-Institutes, Shanghai Tongren Hospital/Faculty of Basic Medicine, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Li Yang
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Chemical Biology Division of Shanghai Universities E-Institutes, Shanghai Tongren Hospital/Faculty of Basic Medicine, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yin Tong
- Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
| | - Xinhua Xiao
- Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
| | - Hu Lei
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Chemical Biology Division of Shanghai Universities E-Institutes, Shanghai Tongren Hospital/Faculty of Basic Medicine, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Wei Liu
- Shanghai First People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
| | - Hanzhang Xu
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Chemical Biology Division of Shanghai Universities E-Institutes, Shanghai Tongren Hospital/Faculty of Basic Medicine, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jiong Deng
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Chemical Biology Division of Shanghai Universities E-Institutes, Shanghai Tongren Hospital/Faculty of Basic Medicine, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Li Zhou
- Department of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin Er Road, Shanghai, China.
| | - Yingli Wu
- Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Chemical Biology Division of Shanghai Universities E-Institutes, Shanghai Tongren Hospital/Faculty of Basic Medicine, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| |
Collapse
|
67
|
Wang XQ, Bai HM, Li ST, Sun H, Min LZ, Tao BB, Zhong J, Li B. Knockdown of HDAC1 expression suppresses invasion and induces apoptosis in glioma cells. Oncotarget 2018. [PMID: 28624794 PMCID: PMC5564623 DOI: 10.18632/oncotarget.18227] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Glioma is the most common malignant tumor of the central nervous system, with a low survival rate of five years worldwide. Although high expression and prognostic value of histone deacetylase 1 (HDAC1) have been recently reported in various types of human tumors, the molecular mechanism underlying the biological function of HDAC1 in glioma is still unclear. We found that HDAC1 was elevated in glioma tissues and cell lines. HDAC1 expression was closely related with pathological grade and overall survival of patients with gliomas. Downregulation of HDAC1 inhibited cell proliferation, prevented invasion of glioma cell lines, and induced cell apoptosis. The expression of apoptosis and metastasis related molecules were detected by RT-PCR and Western blot, respectively, in U251 and T98G cells with HDAC1 knockdown. We found that HDAC1 knockdown upregulated expression of BIM, BAX, cleaved CASPASE3 and E-CADHERIN, and decreased expression of TWIST1, SNAIL and MMP9 in U251 and T98G cells with HDAC1 knockdown. In vivo data showed that knockdown of HDAC1 inhibited tumor growth in nude mice. In summary, HDAC1 may therefore be considered an unfavorable progression indicator for glioma patients, and may also serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Xiao-Qiang Wang
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Hong-Min Bai
- Department of Neurosurgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, China
| | - Shi-Ting Li
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Hui Sun
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ling-Zhao Min
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Bang-Bao Tao
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jun Zhong
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Bin Li
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
68
|
Molecular mechanisms affecting estrogen receptor levels in breast cancer. JOURNAL OF SURGERY AND MEDICINE 2018. [DOI: 10.28982/josam.412314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
69
|
Czaja AJ. Epigenetic changes and their implications in autoimmune hepatitis. Eur J Clin Invest 2018; 48. [PMID: 29383703 DOI: 10.1111/eci.12899] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/25/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND The genetic risk of autoimmune hepatitis is insufficient to explain the observed risk, and epigenetic changes may explain disparities in disease occurrence in different populations within and between countries. The goal of this review was to examine how epigenetic changes induced by the environment or inherited as a phenotypic trait may affect autoimmune hepatitis and be amenable to therapeutic intervention. MATERIALS AND METHODS Pertinent abstracts were identified in PubMed by multiple search terms. The number of abstracts reviewed was 1689, and the number of full-length articles reviewed exceeded 150. RESULTS Activation of pro-inflammatory genes in autoimmune disease is associated with hypomethylation of deoxyribonucleic acid and modification of histones within chromatin. Organ-specific microribonucleic acids can silence genes by marking messenger ribonucleic acids for degradation, and they can promote inflammatory activity or immunosuppression. High circulating levels of the microribonucleic acids 21 and 122 have been demonstrated in autoimmune hepatitis, and they may increase production of pro-inflammatory cytokines. Microribonucleic acids are also essential for maintaining regulatory T cells. Drugs, pollutants, infections, diet and ageing can induce inheritable epigenetic changes favouring autoimmunity. Reversal is feasible by manipulating enzymes, transcription factors, gene-silencing molecules and toxic exposures or by administering methyl donors and correcting vitamin D deficiency. Gene targets, site specificity, efficacy and consequences are uncertain. CONCLUSIONS Potentially reversible epigenetic changes may affect the occurrence and outcome of autoimmune hepatitis, and investigations are warranted to determine the nature of these changes, key genomic targets, and feasible interventions and their consequences.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
70
|
Ibhazehiebo K, Gavrilovici C, de la Hoz CL, Ma SC, Rehak R, Kaushik G, Meza Santoscoy PL, Scott L, Nath N, Kim DY, Rho JM, Kurrasch DM. A novel metabolism-based phenotypic drug discovery platform in zebrafish uncovers HDACs 1 and 3 as a potential combined anti-seizure drug target. Brain 2018; 141:744-761. [PMID: 29373639 PMCID: PMC5837409 DOI: 10.1093/brain/awx364] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 10/29/2017] [Accepted: 11/05/2017] [Indexed: 01/01/2023] Open
Abstract
Despite the development of newer anti-seizure medications over the past 50 years, 30-40% of patients with epilepsy remain refractory to treatment. One explanation for this lack of progress is that the current screening process is largely biased towards transmembrane channels and receptors, and ignores intracellular proteins and enzymes that might serve as efficacious molecular targets. Here, we report the development of a novel drug screening platform that harnesses the power of zebrafish genetics and combines it with in vivo bioenergetics screening assays to uncover therapeutic agents that improve mitochondrial health in diseased animals. By screening commercially available chemical libraries of approved drugs, for which the molecular targets and pathways are well characterized, we were able to reverse-identify the proteins targeted by efficacious compounds and confirm the physiological roles that they play by utilizing other pharmacological ligands. Indeed, using an 870-compound screen in kcna1-morpholino epileptic zebrafish larvae, we uncovered vorinostat (Zolinza™; suberanilohydroxamic acid, SAHA) as a potent anti-seizure agent. We further demonstrated that vorinostat decreased average daily seizures by ∼60% in epileptic Kcna1-null mice using video-EEG recordings. Given that vorinostat is a broad histone deacetylase (HDAC) inhibitor, we then delineated a specific subset of HDACs, namely HDACs 1 and 3, as potential drug targets for future screening. In summary, we have developed a novel phenotypic, metabolism-based experimental therapeutics platform that can be used to identify new molecular targets for future drug discovery in epilepsy.
Collapse
Affiliation(s)
- Kingsley Ibhazehiebo
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Cezar Gavrilovici
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
- Departments of Pediatrics, Clinical Neurosciences, Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Cristiane L de la Hoz
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Shun-Chieh Ma
- Departments of Neurology and Neurobiology, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Renata Rehak
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Gaurav Kaushik
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Paola L Meza Santoscoy
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Lucas Scott
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
- Departments of Pediatrics, Clinical Neurosciences, Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Nandan Nath
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| | - Do-Young Kim
- Departments of Neurology and Neurobiology, Barrow Neurological Institute, St Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Jong M Rho
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
- Departments of Pediatrics, Clinical Neurosciences, Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Canada
| |
Collapse
|
71
|
Histone deacetylase inhibitor ITF2357 leads to apoptosis and enhances doxorubicin cytotoxicity in preclinical models of human sarcoma. Oncogenesis 2018; 7:20. [PMID: 29472530 PMCID: PMC5833676 DOI: 10.1038/s41389-018-0026-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/26/2017] [Accepted: 11/09/2017] [Indexed: 12/14/2022] Open
Abstract
Sarcomas are rare tumors with generally poor prognosis, for which current therapies have shown limited efficacy. Histone deacetylase inhibitors (HDACi) are emerging anti-tumor agents; however, little is known about their effect in sarcomas. By using established and patient-derived sarcoma cells with different subtypes, we showed that the pan-HDACi, ITF2357, potently inhibited in vitro survival in a p53-independent manner. ITF2357-mediated cell death implied the activation of mitochondrial apoptosis, as attested by induction of pro-apoptotic BH3-only proteins and a caspases-dependent mechanism. ITF2357 also induced autophagy, which protected sarcoma cells from apoptotic cell death. ITF2357 activated forkhead box (FOXO) 1 and 3a transcription factors and their downstream target genes, however, silencing of both FOXO1 and 3a did not protect sarcoma cells against ITF2357-induced apoptosis and upregulated FOXO4 and 6. Notably, ITF2357 synergized with Doxorubicin to induce cell death of established and patient-derived sarcoma cells. Furthermore, combination treatment strongly impaired xenograft tumor growth in vivo, when compared to single treatments, suggesting that combination of ITF2357 with Doxorubicin has the potential to enhance sensitization in different preclinical models of sarcoma. Overall, our study highlights the therapeutic potential of ITF2357, alone or in rational combination therapies, for bone and soft tissue sarcomas management.
Collapse
|
72
|
A rapid and simple HPLC–MS/MS method for the simultaneous quantification of valproic acid and its five metabolites in human plasma and application to study pharmacokinetic interaction in Chinese epilepsy patients. J Pharm Biomed Anal 2018; 149:448-456. [DOI: 10.1016/j.jpba.2017.11.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/12/2017] [Accepted: 11/13/2017] [Indexed: 11/21/2022]
|
73
|
Backe MB, Andersson JL, Bacos K, Christensen DP, Hansen JB, Dorosz JJ, Gajhede M, Dahlby T, Bysani M, Kristensen LH, Ling C, Olsen L, Mandrup-Poulsen T. Lysine demethylase inhibition protects pancreatic β cells from apoptosis and improves β-cell function. Mol Cell Endocrinol 2018; 460:47-56. [PMID: 28684291 DOI: 10.1016/j.mce.2017.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 06/27/2017] [Accepted: 07/02/2017] [Indexed: 01/04/2023]
Abstract
Transcriptional changes control β-cell survival in response to inflammatory stress. Posttranslational modifications of histone and non-histone transcriptional regulators activate or repress gene transcription, but the link to cell-fate signaling is unclear. Inhibition of lysine deacetylases (KDACs) protects β cells from cytokine-induced apoptosis and reduces type 1 diabetes incidence in animals. We hypothesized that also lysine demethylases (KDMs) regulate β-cell fate in response to inflammatory stress. Expression of the demethylase Kdm6B was upregulated by proinflammatory cytokines suggesting a possible role in inflammation-induced β-cell destruction. Inhibition of KDM6 demethylases using the selective inhibitor GSK-J4 protected insulin-producing cells and human and mouse islets from cytokine-induced apoptosis by blunting nuclear factor (NF)-κB signaling and endoplasmic reticulum (ER) stress response gene expression. GSK-J4 furthermore increased expression of insulin gene and glucose-stimulated insulin secretion. Expression of genes regulating purinergic and cytokine ligand-receptor interactions was downregulated following GSK-J4 exposure, while expression of genes involved in cell maintenance and survival was upregulated. These data suggest that KDMs are important regulators of inflammation-induced β-cell dysfunction and death.
Collapse
Affiliation(s)
- Marie Balslev Backe
- Immuno-endocrinology Laboratory, Department of Biomedical Sciences, University of Copenhagen, Denmark
| | - Jan Legaard Andersson
- Section of Biostructural Reseach, Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Karl Bacos
- Unit for Epigenetics and Diabetes, Department of Clinical Sciences, Lund University, Scania University Hospital, Malmö, Sweden
| | - Dan Ploug Christensen
- Immuno-endocrinology Laboratory, Department of Biomedical Sciences, University of Copenhagen, Denmark
| | - Jakob Bondo Hansen
- Immuno-endocrinology Laboratory, Department of Biomedical Sciences, University of Copenhagen, Denmark
| | - Jerzy Jòzef Dorosz
- Section of Biostructural Reseach, Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Michael Gajhede
- Section of Biostructural Reseach, Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Tina Dahlby
- Immuno-endocrinology Laboratory, Department of Biomedical Sciences, University of Copenhagen, Denmark
| | - Madhusudhan Bysani
- Unit for Epigenetics and Diabetes, Department of Clinical Sciences, Lund University, Scania University Hospital, Malmö, Sweden
| | - Line Hyltoft Kristensen
- Section of Biostructural Reseach, Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Charlotte Ling
- Unit for Epigenetics and Diabetes, Department of Clinical Sciences, Lund University, Scania University Hospital, Malmö, Sweden
| | - Lars Olsen
- Section of Biostructural Reseach, Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Thomas Mandrup-Poulsen
- Immuno-endocrinology Laboratory, Department of Biomedical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
74
|
Xie Y, Huang L, Chen L, Lin X, Chen L, Zheng Q. Effect of dendritic cell-cytokine-induced killer cells in patients with advanced colorectal cancer combined with first-line treatment. World J Surg Oncol 2017; 15:209. [PMID: 29179719 PMCID: PMC5704402 DOI: 10.1186/s12957-017-1278-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/15/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Surgical resection combined with adjuvant chemotherapy is considered as the gold-standard treatment for advanced colorectal cancer patients. These patients have a poor 5-year survival rate of 5% or less. Furthermore, a large dose of chemotherapy can produce adverse side effects and severe toxicity. Therefore, this retrospective study aimed to evaluate the efficacy of dendritic cell-cytokine-induced killer (DC-CIK) cell infusion as an adjuvant therapy in patients with advanced colorectal cancer combined with first-line treatment. METHODS A total of 142 patients with stage III/IV colorectal carcinoma who had been treated with first-line therapy were included in this study. Among these patients, 71 patients received first-line treatment only (non-DC-CIK group), while the other 71 patients who had similar demographic and clinical characteristics received a DC-CIK cell infusion combined with first-line treatment (DC-CIK group). These patients were followed up until August 2014. Data were analyzed by Kaplan-Meier and Cox regression. RESULTS Our results showed that the 5-year overall survival (OS) rate for the DC-CIK group versus the non-DC-CIK group was 41.3 versus 19.4% (p = 0.001) and the 5-year progression-free survival (PFS) rate for the DC-CIK group versus the non-DC-CIK group was 57.4 versus 33.6% (p = 0.022). CONCLUSIONS Our results showed that patients with advanced colorectal cancer might benefit from DC-CIK immunotherapy combined with first-line therapy by significantly prolonging 5-year OS and PFS.
Collapse
Affiliation(s)
- Yunqing Xie
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Lijie Huang
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Luchuan Chen
- Department of Abdominal Surgery, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Xiaowei Lin
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Li Chen
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Qiuhong Zheng
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China.
| |
Collapse
|
75
|
Li YZ, Yu S, Yan PA, Gong DY, Wu FL, He Z, Yuan YY, Zhao AY, Tang X, Zhang RQ, Peng C, Cao ZX. Crotonoside exhibits selective post-inhibition effect in AML cells via inhibition of FLT3 and HDAC3/6. Oncotarget 2017; 8:103087-103099. [PMID: 29262547 PMCID: PMC5732713 DOI: 10.18632/oncotarget.20710] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 08/02/2017] [Indexed: 12/20/2022] Open
Abstract
Targeted therapies for the treatment of acute myeloid leukemia (AML), specifically the FLT3 inhibitors, have shown promising results. Nevertheless, it is very unlikely that inhibitors which target a single pathway will provide long-term disease control. Here, we report the characterization of crotonoside, a natural product extracted from Chinese medicinal herb, Croton, for the treatment of AML via inhibition of FLT3 and HDAC3/6. In vitro, crotonoside exhibited selective inhibition in AML cells. In vivo, crotonoside treatment at 70 and 35 mg/kg/d produced significant AML tumor inhibition rates of 93.5% and 73.6%, respectively. Studies on the anti-AML mechanism of crotonoside demonstrated a significant inhibition of FLT3 signaling, cell cycle arrest in G0/G1 phase, and apoptosis. In contrast to classic FLT3 inhibitor; sunitinib, crotonoside was able to selectively suppress the expression of HDAC3 and HDAC6 without altering the expression of other HDAC isoforms. Inhibitors of HDAC3 and HDAC6; RGFP966 and HPOB, respectively, also exhibited selective inhibition in AML cells. Furthermore, we established novel signaling pathways including HDAC3/NF-κB-p65 and HDAC6/c-Myc besides FLT3/c-Myc which are aberrantly regulated in the progression of AML. In addition, crotonoside alone or the combination of sunitinib/RFP966/HPOB exhibited a significant post-inhibition effect in AML cells by the inhibition of FLT3 and HDAC3/6. Inhibitors targeting the FLT3 and HDAC3/6 might provide a more effective treatment strategy for AML. Taken together, the present study suggests that crotonoside could be a promising candidate for the treatment of AML, and deserves further investigations.
Collapse
Affiliation(s)
- Yu-Zhi Li
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province-Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, Sichuan, China
| | - Si Yu
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province-Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, Sichuan, China
| | - Pei-Ao Yan
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province-Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, Sichuan, China
| | - Dao-Yin Gong
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province-Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, Sichuan, China
| | - Fang-Li Wu
- Second Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Zhi He
- Second Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Yu-Yao Yuan
- Second Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - An-Yan Zhao
- Second Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Sichuan, China
| | - Xue Tang
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province-Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, Sichuan, China
| | - Ruo-Qi Zhang
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province-Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, Sichuan, China
| | - Cheng Peng
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province-Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, Sichuan, China
| | - Zhi-Xing Cao
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicine, Key Laboratory of Systematic Research, Development and Utilization of Chinese Medicine Resources in Sichuan Province-Key Laboratory Breeding Base of Co-founded by Sichuan Province and MOST, Sichuan, China
| |
Collapse
|
76
|
Androutsopoulos VP, Spandidos DA. Antiproliferative effects of TSA, PXD‑101 and MS‑275 in A2780 and MCF7 cells: Acetylated histone H4 and acetylated tubulin as markers for HDACi potency and selectivity. Oncol Rep 2017; 38:3412-3418. [PMID: 29039546 PMCID: PMC5783587 DOI: 10.3892/or.2017.6015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 10/03/2017] [Indexed: 12/26/2022] Open
Abstract
Inhibition of histone deacetylase enzymes (HDACs) has been well documented as an attractive target for the development of chemotherapeutic drugs. The present study investigated the effects of two prototype hydroxamic acid HDAC inhibitors, namely Trichostatin A (TSA) and Belinostat (PXD-101) and the benzamide Entinostat (MS-275) in A2780 ovarian carcinoma and MCF7 breast adenocarcinoma cells. The three HDACi inhibited the proliferation of A2780 and MCF7 cells at comparable levels, below the µM range. Enzyme inhibition assays in a cell-free system showed that TSA was the most potent inhibitor of total HDAC enzyme activity followed by PXD-101 and MS-275. Incubation of A2780 and MCF7 cells with the hydroxamates TSA and PXD-101 for 24 h resulted in a dramatic increase of acetylated tubulin induction (up to 30-fold for TSA). In contrast to acetylated tubulin, western blot analysis and flow cytometry indicated that the induction of acetylated histone H4 was considerably smaller. The benzamide MS-275 exhibited nearly a 2-fold induction of acetylated histone H4 and an even smaller induction of acetylated tubulin in A2780 and MCF7 cells. Taken together, these data suggest that although the three HDACi were equipotent in inhibiting proliferation of MCF7 and A2780 cells, only the benzamide MS-275 did not induce acetylated tubulin expression, a marker of class IIb HDACs.
Collapse
Affiliation(s)
- Vasilis P Androutsopoulos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, Voutes 71003, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, Voutes 71003, Greece
| |
Collapse
|
77
|
Moyal L, Goldfeiz N, Gorovitz B, Rephaeli A, Tal E, Tarasenko N, Nudelman A, Ziv Y, Hodak E. AN-7, a butyric acid prodrug, sensitizes cutaneous T-cell lymphoma cell lines to doxorubicin via inhibition of DNA double strand breaks repair. Invest New Drugs 2017; 36:1-9. [PMID: 28884410 DOI: 10.1007/s10637-017-0500-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/10/2017] [Indexed: 12/13/2022]
Abstract
We previously found that the novel histone deacetylase inhibitor (HDACI) butyroyloxymethyl diethylphosphate (AN-7) had greater selectivity against cutaneous T-cell lymphoma (CTCL) than SAHA. AN-7 synergizes with doxorubicin (Dox), an anthracycline antibiotic that induces DNA breaks. This study aimed to elucidate the mechanism underlying the effect of AN-7 on Dox-induced double-strand DNA breaks (DSBs) in CTCL, MyLa and Hut78 cell lines. The following markers/assays were employed: comet assay; western blot of γH2AX and p-KAP1; immunofluorescence of γH2AX nuclear foci; Western blot of repair protein; quantification of DSBs-repair through homologous recombination. DSB induction by Dox was evidenced by an increase in DSB markers, and DSBs-repair, by their subsequent decrease. The addition of AN-7 slightly increased Dox induction of DSBs in MyLa cells with no effect in Hut78 cells. AN-7 inhibited the repair of Dox-induced DSBs, with a more robust effect in Hut78. Treatment with AN-7 followed by Dox reduced the expression of DSB-repair proteins, with direct interference of AN-7 with the homologous recombination repair. AN-7 sensitizes CTCL cell lines to Dox, and when combined with Dox, sustains unrepaired DSBs by suppressing repair protein expression. Our data provide a mechanistic rationale for combining AN-7 with Dox or other DSB inducers as a therapeutic modality in CTCL.
Collapse
Affiliation(s)
- Lilach Moyal
- Laboratory for Molecular Dermatology, Felsenstein Medical Research Center, Petach Tikva and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Department of Dermatology, Rabin Medical Center, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Neta Goldfeiz
- Laboratory for Molecular Dermatology, Felsenstein Medical Research Center, Petach Tikva and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Dermatology, Rabin Medical Center, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Batia Gorovitz
- Laboratory for Molecular Dermatology, Felsenstein Medical Research Center, Petach Tikva and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Dermatology, Rabin Medical Center, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ada Rephaeli
- Laboratory for Pharmacology and Experimental Oncology, Felsenstein Medical Research Center, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Efrat Tal
- The David and Inez Myers Laboratory for Cancer Research, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nataly Tarasenko
- Laboratory for Pharmacology and Experimental Oncology, Felsenstein Medical Research Center, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Abraham Nudelman
- Division of Medicinal Chemistry, Department of Chemistry, Bar Ilan University, Ramat Gan, Israel
| | - Yael Ziv
- The David and Inez Myers Laboratory for Cancer Research, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Emmilia Hodak
- Laboratory for Molecular Dermatology, Felsenstein Medical Research Center, Petach Tikva and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Dermatology, Rabin Medical Center, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
78
|
Histone Deacetylase Inhibitor-Induced Autophagy in Tumor Cells: Implications for p53. Int J Mol Sci 2017; 18:ijms18091883. [PMID: 30563957 PMCID: PMC5618532 DOI: 10.3390/ijms18091883] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/20/2017] [Accepted: 08/28/2017] [Indexed: 02/07/2023] Open
Abstract
Autophagy is an essential process of the eukaryotic cell allowing degradation and recycling of dysfunctional cellular components in response to either physiological or pathological changes. Inhibition of autophagy in combination with chemotherapeutic treatment has emerged as a novel approach in cancer treatment leading to cell cycle arrest, differentiation, and apoptosis. Suberoyl hydroxamic acid (SAHA) is a broad-spectrum histone deacetylase inhibitor (HDACi) suppressing family members in multiple HDAC classes. Increasing evidence indicates that SAHA and other HDACi can, in addition to mitochondria-mediated apoptosis, also promote caspase-independent autophagy. SAHA-induced mTOR inactivation as a major regulator of autophagy activating the remaining autophagic core machinery is by far the most reported pathway in several tumor models. However, the question of which upstream mechanisms regulate SAHA-induced mTOR inactivation that consequently initiate autophagy has been mainly left unexplored. To elucidate this issue, we recently initiated a study clarifying different modes of SAHA-induced cell death in two human uterine sarcoma cell lines which led to the conclusion that the tumor suppressor protein p53 could act as a molecular switch between SAHA-triggered autophagic or apoptotic cell death. In this review, we present current research evidence about HDACi-mediated apoptotic and autophagic pathways, in particular with regard to p53 and its therapeutic implications.
Collapse
|
79
|
Duvic M, Bates SE, Piekarz R, Eisch R, Kim YH, Lerner A, Robak T, Samtsov A, Becker JC, McCulloch W, Waksman J, Whittaker S. Responses to romidepsin in patients with cutaneous T-cell lymphoma and prior treatment with systemic chemotherapy. Leuk Lymphoma 2017; 59:880-887. [PMID: 28853310 DOI: 10.1080/10428194.2017.1361022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cutaneous T-cell lymphomas (CTCL) are a group of non-Hodgkin lymphomas that typically present in the skin but can progress to systemic involvement. The optimal treatment for patients who relapse from or are refractory to systemic chemotherapy remains unclear. Romidepsin is a potent, class-I selective histone deacetylase inhibitor approved for the treatment of patients with CTCL who have had ≥1 prior systemic therapy. Here, we present a subanalysis of two phase-2 trials (NCT00106431, NCT00007345) of romidepsin in patients with CTCL who had prior treatment with systemic chemotherapy. Patients with prior chemotherapy were able to achieve durable responses to romidepsin, and response rates were similar to those in patients who were chemotherapy naïve. Overall, no new safety signals emerged in patients who had received prior chemotherapy. The data presented here suggest that romidepsin is safe and effective in patients with CTCL who received prior systemic chemotherapy.
Collapse
Affiliation(s)
- Madeleine Duvic
- a The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Susan E Bates
- b Department of Hematology/Oncology , Columbia University , New York , NY , USA
| | - Richard Piekarz
- c Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis , National Cancer Institute , Bethesda , MD , USA
| | - Robin Eisch
- d National Cancer Institute , Bethesda , MD , USA
| | - Youn H Kim
- e Department of Dermatology , Stanford University , Stanford , CA , USA
| | | | - Tadeusz Robak
- g Department of Hematology , Medical University of Lodz, Copernicus Memorial Hospital , Lodz , Poland
| | - Alexey Samtsov
- h State Educational Institution of Military Medicine , St. Petersburg , Russia
| | - Jürgen C Becker
- i Translational Skin Cancer Research, German Cancer Consortium (DKTK) , University Clinic Essen , Germany
| | | | - Joel Waksman
- k Brightech International LLC , Somerset , NJ , USA
| | | |
Collapse
|
80
|
Kumar N, Tomar R, Pandey A, Tomar V, Singh VK, Chandra R. Preclinical evaluation and molecular docking of 1,3-benzodioxole propargyl ether derivatives as novel inhibitor for combating the histone deacetylase enzyme in cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1288-1299. [PMID: 28847179 DOI: 10.1080/21691401.2017.1369423] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Even after huge strides in medicine, cancer continues to be a formidable disease, which is slated to become the leading cause of death worldwide. The present study investigates the 1,3-benzodioxole and its propargyl ether derivatives as a novel histone deacetylase enzyme inhibitor in order to cure cancer, as aberrant expression of histone deacetylases (HDACs) is associated with carcinogenesis. Bioinformatics approaches were employed to carry out preclinical and pharmacological evaluations of designed benzodioxole derivatives. Furthermore, their interaction with HDAC-1 enzyme was studied through computational methods for their specific inhibitory effects and evaluated for their LD50 (oral rat acute toxicity) value. In addition to this work, three-dimensional (3D) structure of HDAC-1 enzyme was extracted and evaluated using various parameters including Ramachandran plot and molecular docking stimulation. In our study, we found that compound 7 and compound 9 have higher binding score than approved drugs (SAHA, TSA and VPA). Importantly, these compounds were found to possess good pharmacological and pharmacokinetic properties and can be considered as potent novel compound to combat the HDAC-1 enzyme to cure cancer. Compounds were also analyzed and validated with parameters like absorption, metabolism, excretion, toxicity and synthetic accessibility during the preclinical evaluation. This study paves way to search for novel and potent small chemical compounds for inhibiting HDAC-1 enzyme and in particular to combat the cancer progression by interrupting the cell cycle.
Collapse
Affiliation(s)
- Neeraj Kumar
- a Department of Chemistry, Drug Discovery and Development Laboratory , University of Delhi , Delhi , India.,b Department of Biotechnology, Stem Cell Research Laboratory , Delhi Technological University , Delhi , India
| | - Ravi Tomar
- a Department of Chemistry, Drug Discovery and Development Laboratory , University of Delhi , Delhi , India
| | - Apurva Pandey
- a Department of Chemistry, Drug Discovery and Development Laboratory , University of Delhi , Delhi , India
| | - Vartika Tomar
- a Department of Chemistry, Drug Discovery and Development Laboratory , University of Delhi , Delhi , India
| | - Vimal Kishor Singh
- c Dr. B. R. Ambedkar Centre for Biomedical Research , University of Delhi , Delhi , India
| | - Ramesh Chandra
- a Department of Chemistry, Drug Discovery and Development Laboratory , University of Delhi , Delhi , India.,c Dr. B. R. Ambedkar Centre for Biomedical Research , University of Delhi , Delhi , India
| |
Collapse
|
81
|
Liu J, Wang T, Wang X, Luo L, Guo J, Peng Y, Xu Q, Miao J, Zhang Y, Ling Y. Development of novel β-carboline-based hydroxamate derivatives as HDAC inhibitors with DNA damage and apoptosis inducing abilities. MEDCHEMCOMM 2017; 8:1213-1219. [PMID: 30108831 DOI: 10.1039/c6md00681g] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/22/2017] [Indexed: 12/22/2022]
Abstract
A series of novel β-carboline-based hydroxamate derivatives (8a-n) as HDAC inhibitors have been designed and synthesized. Most of these compounds displayed potent histone deacetylase inhibitory effects and good antiproliferative activity with IC50s in the low micromolar range. One of the most potent compounds (8k) showed the strongest inhibition of the proliferation of human hepatocellular carcinoma (HCC) cells in vitro, with IC50 values lower than that of the currently approved HDAC inhibitor SAHA. Compound 8k also increased acetylation of histone H3 and α-tubulin, consistent with its potent HDAC inhibition. Importantly, 8k induced hypochromism by electrostatic interactions with CT-DNA, suggesting potential induction of DNA damage. Finally, 8k significantly induced HepG2 cell apoptosis by regulating apoptotic relative proteins expression. Together, our findings suggest that these novel β-carboline-based hydroxamate derivatives may provide a new framework for the discovery of novel antitumor agents for the intervention of human carcinoma cells.
Collapse
Affiliation(s)
- Ji Liu
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target , Nantong University , Nantong 226001 , P.R. China . ;
| | - Tingting Wang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target , Nantong University , Nantong 226001 , P.R. China . ;
| | - Xinyang Wang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target , Nantong University , Nantong 226001 , P.R. China . ; .,State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing 210009 , P.R. China
| | - Lin Luo
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target , Nantong University , Nantong 226001 , P.R. China . ;
| | - Jing Guo
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target , Nantong University , Nantong 226001 , P.R. China . ;
| | - Yanfu Peng
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target , Nantong University , Nantong 226001 , P.R. China . ;
| | - Qibing Xu
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target , Nantong University , Nantong 226001 , P.R. China . ;
| | - Jiefei Miao
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target , Nantong University , Nantong 226001 , P.R. China . ; .,Tumor-chemotherapy Department , Affiliated Hospital , Nantong University , Nantong 226001 , P.R. China
| | - Yanan Zhang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target , Nantong University , Nantong 226001 , P.R. China . ;
| | - Yong Ling
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target , Nantong University , Nantong 226001 , P.R. China . ; .,State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing 210009 , P.R. China
| |
Collapse
|
82
|
van Gucht ALM, Moran C, Meima ME, Visser WE, Chatterjee K, Visser TJ, Peeters RP. Resistance to Thyroid Hormone due to Heterozygous Mutations in Thyroid Hormone Receptor Alpha. Curr Top Dev Biol 2017; 125:337-355. [PMID: 28527577 DOI: 10.1016/bs.ctdb.2017.02.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND Thyroid hormone (TH) acts via nuclear thyroid hormone receptors (TRs). TR isoforms (TRα1, TRα2, TRβ1, TRβ2) are encoded by distinct genes (THRA and THRB) and show differing tissue distributions. Patients with mutations in THRB, exhibiting resistance within the hypothalamic-pituitary-thyroid axis with elevated TH and nonsuppressed thyroid-stimulating hormone (TSH) levels, were first described decades ago. In 2012, the first patients with mutations in THRA were identified. Scope of this review: This review describes the clinical and biochemical characteristics of patients with resistance to thyroid hormone alpha (RTHα) due to heterozygous mutations in THRA. The genetic basis and molecular pathogenesis of the disorder together with effects of levothyroxine treatment are discussed. CONCLUSIONS The severity of the clinical phenotype of RTHα patients seems to be associated with the location and type of mutation in THRA. The most frequent abnormalities observed include anemia, constipation, and growth and developmental delay. In addition, serum (F)T3 levels can be high-normal to high, (F)T4 and rT3 levels normal to low, while TSH is normal or mildly raised. Despite heterogeneous consequences of mutations in THRA, RTHα should be suspected in subjects with even mild clinical features of hypothyroidism together with high/high-normal (F)T3, low/low-normal (F)T4, and normal TSH.
Collapse
Affiliation(s)
| | - Carla Moran
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Marcel E Meima
- Erasmus University Medical Center, Rotterdam, The Netherlands
| | - W Edward Visser
- Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Krishna Chatterjee
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Theo J Visser
- Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Robin P Peeters
- Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
83
|
Hailu GS, Robaa D, Forgione M, Sippl W, Rotili D, Mai A. Lysine Deacetylase Inhibitors in Parasites: Past, Present, and Future Perspectives. J Med Chem 2017; 60:4780-4804. [DOI: 10.1021/acs.jmedchem.6b01595] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Gebremedhin S. Hailu
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
| | - Dina Robaa
- Institute of Pharmacy, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Mariantonietta Forgione
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
- Center
for Life Nano Science@Sapienza, Italian Institute of Technology, Viale Regina Elena 291, 00161 Rome, Italy
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther-Universitat Halle-Wittenberg, Halle, Germany
| | - Dante Rotili
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
| | - Antonello Mai
- Dipartimento
di Chimica e Tecnologie del Farmaco “Sapienza” Università di Roma, 00185 Rome, Italy
- Istituto
Pasteur, Fondazione Cenci-Bolognetti, “Sapienza” Università di Roma, 00185 Rome, Italy
| |
Collapse
|
84
|
Um S, Lee H, Zhang Q, Kim HY, Lee JH, Seo BM. Valproic Acid Modulates the Multipotency in Periodontal Ligament Stem Cells via p53-Mediated Cell Cycle. Tissue Eng Regen Med 2017; 14:153-162. [PMID: 30603472 DOI: 10.1007/s13770-017-0027-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 07/31/2016] [Accepted: 08/04/2016] [Indexed: 12/20/2022] Open
Abstract
Human periodontal ligament stem cells (PDLSCs), a type of mesenchymal stem cell, are a promising source for dental regeneration and are identified in human periodontal ligaments from extracted third molars. Valproic acid (VPA) is a histone deacetylase inhibitor that has been used as a wide-spectrum antiepileptic drug and a medication for mood disorders. VPA has shown several effects on increasing the pluripotency of embryonic stem cells and controlling osteogenic differentiation, besides the prevention of seizures. However, its effect on proliferation and osteogenesis depends on the cell type and concentration. The aim of this study was to investigate the effects of cyclic and constant VPA treatment on PDLSCs. Proliferation and apoptosis of PDLSCs were determined with cyclic and constant VPA treatment. In cemento/osteogenic differentiation, osteogenic markers decreased significantly after cyclic treatment with 0.5 mM VPA. In contrast, VPA enhanced osteogenic differentiation after constant treatment. With cyclic VPA treatment, p53 levels related to apoptotic pathway decreased to induce proliferation. These findings indicated that VPA has different roles in proliferation and differentiation of PDLSCs in vitro and in vivo via p53-related pathway.
Collapse
Affiliation(s)
- Soyoun Um
- 1Department of Dental Science, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- 5Dental Research Institute, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Ho Lee
- 2Department of Oral and Maxillofacial Surgery, SMG-SNU Boramae Medical Center, Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061 Korea
| | - Qingbin Zhang
- 3Department of Temporomandibular Joint Diseases, School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou, 510182 China
| | - Hui Young Kim
- 4Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- 5Dental Research Institute, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Joo-Hee Lee
- 4Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- 5Dental Research Institute, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Byoung Moo Seo
- 4Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
- 5Dental Research Institute, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| |
Collapse
|
85
|
Shustov A, Coiffier B, Horwitz S, Sokol L, Pro B, Wolfson J, Balser B, Eisch R, Popplewell L, Prince HM, Allen SL, Piekarz R, Bates S. Romidepsin is effective and well tolerated in older patients with peripheral T-cell lymphoma: analysis of two phase II trials. Leuk Lymphoma 2017; 58:2335-2341. [PMID: 28264616 DOI: 10.1080/10428194.2017.1295143] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Peripheral T-cell lymphomas (PTCLs) are a rare group of lymphoid neoplasms with high relapse rates after initial therapy and poor prognosis. Most patients are aged ≥60 years and are often not candidates for aggressive salvage therapies. Romidepsin, a potent class I histone deacetylase inhibitor, has shown significant single-agent activity in relapsed/refractory PTCL. We evaluated the efficacy and tolerability of romidepsin in elderly patients in this setting. Ninety-five patients aged ≥60 years were identified from 2 prospective phase 2 registration trials of romidepsin, and comparative analyses were performed with younger patients from these trials. Response rates, progression-free survival, and overall survival were not statistically different for younger vs older patients. The toxicity profile in older and younger patients was similar in both trials. Romidepsin demonstrated similar efficacy and tolerability in younger and older patients and presents an attractive treatment option for relapsed/refractory PTCL regardless of age. TRIAL REGISTRATION Clinicaltrials.gov identifiers: NCT00426764, NCT00007345.
Collapse
Affiliation(s)
- Andrei Shustov
- a Department of Hematology , University of Washington Medical Center , Seattle , WA , USA
| | - Bertrand Coiffier
- b Department of Hematology , Hospices Civils de Lyon , Lyon , France
| | - Steven Horwitz
- c Department of Oncology , Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Lubomir Sokol
- d Department of Hematology , Moffitt Cancer Center , Tampa , FL , USA
| | - Barbara Pro
- e Department of Hematology/Oncology , Kimmel Cancer Center, Thomas Jefferson University , Philadelphia , PA , USA
| | | | | | - Robin Eisch
- g Department of Investigational Therapeutics , National Cancer Institute , Bethesda , MD , USA
| | - Leslie Popplewell
- h Department of Hematology/Oncology , City of Hope Medical Center-Oncology , Duarte , CA , USA
| | - H Miles Prince
- i Department of Hematology , Peter MacCallum Cancer Centre and University of Melbourne , Melbourne , Australia
| | - Steven L Allen
- j Department of Hematology , Hofstra North Shore-LIJ School of Medicine and Monter Cancer Center , Lake Success , NY , USA
| | - Richard Piekarz
- g Department of Investigational Therapeutics , National Cancer Institute , Bethesda , MD , USA
| | - Susan Bates
- k Department of Hematology/Oncology , Columbia University Medical Center , New York , NY , USA
| |
Collapse
|
86
|
Hrgovic I, Doll M, Pinter A, Kaufmann R, Kippenberger S, Meissner M. Histone deacetylase inhibitors interfere with angiogenesis by decreasing endothelial VEGFR-2 protein half-life in part via a VE-cadherin-dependent mechanism. Exp Dermatol 2017; 26:194-201. [DOI: 10.1111/exd.13159] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Igor Hrgovic
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Monika Doll
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Andreas Pinter
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Roland Kaufmann
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Stefan Kippenberger
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Markus Meissner
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| |
Collapse
|
87
|
Castillo-Aguilera O, Depreux P, Halby L, Arimondo PB, Goossens L. DNA Methylation Targeting: The DNMT/HMT Crosstalk Challenge. Biomolecules 2017; 7:biom7010003. [PMID: 28067760 PMCID: PMC5372715 DOI: 10.3390/biom7010003] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/08/2016] [Accepted: 12/12/2016] [Indexed: 12/22/2022] Open
Abstract
Chromatin can adopt a decondensed state linked to gene transcription (euchromatin) and a condensed state linked to transcriptional repression (heterochromatin). These states are controlled by epigenetic modulators that are active on either the DNA or the histones and are tightly associated to each other. Methylation of both DNA and histones is involved in either the activation or silencing of genes and their crosstalk. Since DNA/histone methylation patterns are altered in cancers, molecules that target these modifications are interesting therapeutic tools. We present herein a vast panel of DNA methyltransferase inhibitors classified according to their mechanism, as well as selected histone methyltransferase inhibitors sharing a common mode of action.
Collapse
Affiliation(s)
- Omar Castillo-Aguilera
- Univ. Lille, ICPAL, EA 7365-GRITA-Groupe de Recherche sur les formes Injectables et les Technologies Associées, 3 rue du Pr. Laguesse, F-59000 Lille, France.
| | - Patrick Depreux
- Univ. Lille, ICPAL, EA 7365-GRITA-Groupe de Recherche sur les formes Injectables et les Technologies Associées, 3 rue du Pr. Laguesse, F-59000 Lille, France.
| | - Ludovic Halby
- FRE3600 Epigenetic Targeting of Cancer, CNRS, 31035 Toulouse, France.
| | - Paola B Arimondo
- FRE3600 Epigenetic Targeting of Cancer, CNRS, 31035 Toulouse, France.
- Churchill College, Cambridge CB3 0DS, UK.
| | - Laurence Goossens
- Univ. Lille, ICPAL, EA 7365-GRITA-Groupe de Recherche sur les formes Injectables et les Technologies Associées, 3 rue du Pr. Laguesse, F-59000 Lille, France.
| |
Collapse
|
88
|
Wu Y, Dai W, Chen X, Geng A, Chen Y, Lu T, Zhu Y. Design, synthesis and biological evaluation of 2,3-dihydroimidazo[1,2-c]quinazoline derivatives as novel phosphatidylinositol 3-kinase and histone deacetylase dual inhibitors. RSC Adv 2017. [DOI: 10.1039/c7ra08835c] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A novel design approach by combination of PI3K and HDAC inhibitory activity in one molecule to produce dual inhibitors.
Collapse
Affiliation(s)
- Yichao Wu
- Department of Organic Chemistry
- School of Science
- China Pharmaceutical University
- Nanjing 211198
- China
| | - Weichen Dai
- Department of Organic Chemistry
- School of Science
- China Pharmaceutical University
- Nanjing 211198
- China
| | - Xin Chen
- Department of Organic Chemistry
- School of Science
- China Pharmaceutical University
- Nanjing 211198
- China
| | - Aixin Geng
- Department of Organic Chemistry
- School of Science
- China Pharmaceutical University
- Nanjing 211198
- China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery
- School of Science
- China Pharmaceutical University
- Nanjing 211198
- China
| | - Tao Lu
- Department of Organic Chemistry
- School of Science
- China Pharmaceutical University
- Nanjing 211198
- China
| | - Yong Zhu
- Department of Organic Chemistry
- School of Science
- China Pharmaceutical University
- Nanjing 211198
- China
| |
Collapse
|
89
|
Hessmann E, Johnsen SA, Siveke JT, Ellenrieder V. Epigenetic treatment of pancreatic cancer: is there a therapeutic perspective on the horizon? Gut 2017; 66:168-179. [PMID: 27811314 PMCID: PMC5256386 DOI: 10.1136/gutjnl-2016-312539] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/30/2016] [Indexed: 12/24/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) constitutes one of the most aggressive malignancies with a 5-year survival rate of <7%. Due to growing incidence, late diagnosis and insufficient treatment options, PDAC is predicted to soon become one of the leading causes of cancer-related death. Although intensified cytostatic combinations, particularly gemcitabine plus nab-paclitaxel and the folinic acid, fluorouracil, irinotecan, oxaliplatin (FOLFIRINOX) protocol, provide some improvement in efficacy and survival compared with gemcitabine alone, a breakthrough in the treatment of metastatic pancreatic cancer remains out of sight. Nevertheless, recent translational research activities propose that either modulation of the immune response or pharmacological targeting of epigenetic modifications alone, or in combination with chemotherapy, might open highly powerful therapeutic avenues in GI cancer entities, including pancreatic cancer. Deregulation of key epigenetic factors and chromatin-modifying proteins, particularly those responsible for the addition, removal or recognition of post-translational histone modifications, are frequently found in human pancreatic cancer and hence constitute particularly exciting treatment opportunities. This review summarises both current clinical trial activities and discovery programmes initiated throughout the biopharma landscape, and critically discusses the chances, hurdles and limitations of epigenetic-based therapy in future PDAC treatment.
Collapse
Affiliation(s)
- Elisabeth Hessmann
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Steven A Johnsen
- Department of General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Goettingen, Germany
| | - Jens T Siveke
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany,West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
| |
Collapse
|
90
|
Foss F, Pro B, Miles Prince H, Sokol L, Caballero D, Horwitz S, Coiffier B. Responses to romidepsin by line of therapy in patients with relapsed or refractory peripheral T-cell lymphoma. Cancer Med 2017; 6:36-44. [PMID: 27981793 PMCID: PMC5269566 DOI: 10.1002/cam4.939] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/21/2016] [Accepted: 09/23/2016] [Indexed: 01/23/2023] Open
Abstract
Peripheral T-cell lymphoma (PTCL) is a heterogeneous group of aggressive non-Hodgkin lymphomas typically associated with poor prognosis. Most patients with PTCL receive chemotherapy as first-line treatment, but many experience rapid relapse. For patients with relapsed/refractory PTCL, responses to treatment and long-term outcomes tend to worsen with increasing lines of therapy. Romidepsin is a potent class I histone deacetylase inhibitor approved by the US Food and Drug Administration for the treatment of PTCL in patients who have received ≥1 prior therapy. A pivotal phase 2 trial of romidepsin in patients with relapsed/refractory PTCL demonstrated an objective response rate of 25% (33/130), including 15% with confirmed/unconfirmed complete response, and a median duration of response of 28 months. In the analysis presented herein, romidepsin was shown to have similar responses and long-term outcomes in patients with 1, 2, and ≥3 prior lines of treatment, including in patients with disease refractory to the last prior therapy. Although adverse events increased with increasing lines of treatment, the rate of dose modifications and discontinuations due to adverse events was not significantly different. These data support the use of romidepsin as salvage treatment for PTCL irrespective of the number of prior therapies.
Collapse
Affiliation(s)
| | - Barbara Pro
- Thomas Jefferson UniversityPhiladelphiaPennsylvania
| | - H. Miles Prince
- Peter MacCallum Cancer CentreUniversity of MelbourneMelbourne, VictoriaAustralia
| | | | | | | | | |
Collapse
|
91
|
Brevini TAL, Pennarossa G, Manzoni EFM, Gandolfi CE, Zenobi A, Gandolfi F. The quest for an effective and safe personalized cell therapy using epigenetic tools. Clin Epigenetics 2016; 8:119. [PMID: 27891192 PMCID: PMC5112765 DOI: 10.1186/s13148-016-0283-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 11/02/2016] [Indexed: 11/10/2022] Open
Abstract
In the presence of different environmental cues that are able to trigger specific responses, a given genotype has the ability to originate a variety of different phenotypes. This property is defined as plasticity and allows cell fate definition and tissue specialization. Fundamental epigenetic mechanisms drive these modifications in gene expression and include DNA methylation, histone modifications, chromatin remodeling, and microRNAs. Understanding these mechanisms can provide powerful tools to switch cell phenotype and implement cell therapy. Environmentally influenced epigenetic changes have also been associated to many diseases such as cancer and neurodegenerative disorders, with patients that do not respond, or only poorly respond, to conventional therapy. It is clear that disorders based on an individual's personal genomic/epigenomic profile can rarely be successfully treated with standard therapies due to genetic heterogeneity and epigenetic alterations and a personalized medicine approach is far more appropriate to manage these patients. We here discuss the recent advances in small molecule approaches for personalized medicine, drug targeting, and generation of new cells for medical application. We also provide prospective views of the possibility to directly convert one cell type into another, in a safe and robust way, for cell-based clinical trials and regenerative medicine.
Collapse
Affiliation(s)
- T A L Brevini
- Laboratory of Biomedical Embryology, Unistem, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| | - G Pennarossa
- Laboratory of Biomedical Embryology, Unistem, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| | - E F M Manzoni
- Laboratory of Biomedical Embryology, Unistem, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| | - C E Gandolfi
- Laboratory of Biomedical Embryology, Unistem, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| | - A Zenobi
- Laboratory of Biomedical Embryology, Unistem, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| | - F Gandolfi
- Laboratory of Biomedical Embryology, Unistem, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| |
Collapse
|
92
|
Foss F, Duvic M, Lerner A, Waksman J, Whittaker S. Clinical Efficacy of Romidepsin in Tumor Stage and Folliculotropic Mycosis Fungoides. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2016; 16:637-643. [DOI: 10.1016/j.clml.2016.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/28/2016] [Accepted: 08/02/2016] [Indexed: 10/21/2022]
|
93
|
Irlé C, Weintraub J. Long-Term Treatment with Romidepsin in Patients with Peripheral T-Cell Lymphoma. Case Rep Hematol 2016; 2016:8175957. [PMID: 27648317 PMCID: PMC5014944 DOI: 10.1155/2016/8175957] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 07/22/2016] [Accepted: 08/02/2016] [Indexed: 12/03/2022] Open
Abstract
Peripheral T-cell lymphomas (PTCLs) are a heterogeneous group of aggressive non-Hodgkin lymphomas. Angioimmunoblastic T-cell lymphoma (AITL) is a common subtype of PTCL, and patients with AITL typically have a poor prognosis with limited treatment options. Clinical studies have demonstrated the activity of romidepsin, a structurally unique, potent, bicyclic class 1 selective histone deacetylase inhibitor, in patients with relapsed or refractory AITL. In the case presented herein, we describe a patient treated with single-agent romidepsin at first diagnosis of AITL, resulting in complete remission for over 2 years and leading to the use of maintenance dosing. The patient eventually underwent a successful autologous stem cell transplant. This case illustrates the successful use of romidepsin for the long-term treatment of a patient with AITL in a clinical setting. Maintenance dosing may be an option for patients who have an extended response to romidepsin in order to optimize outcomes and to prolong time to the next subsequent line of therapy. In our case, the patient was able to remain in complete remission for more than 1 year while receiving maintenance dosing of romidepsin.
Collapse
Affiliation(s)
- Claudius Irlé
- Hôpital de la Tour Unite d'Oncologie-Hematologie, Meyrin, Switzerland
| | | |
Collapse
|
94
|
Morandini AC, Santos CF, Yilmaz Ö. Role of epigenetics in modulation of immune response at the junction of host-pathogen interaction and danger molecule signaling. Pathog Dis 2016; 74:ftw082. [PMID: 27542389 DOI: 10.1093/femspd/ftw082] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2016] [Indexed: 12/17/2022] Open
Abstract
Epigenetic mechanisms have rapidly and controversially emerged as silent modulators of host defenses that can lead to a more prominent immune response and shape the course of inflammation in the host. Thus, the epigenetics can both drive the production of specific inflammatory mediators and control the magnitude of the host response. The epigenetic actions that are predominantly shown to modulate the host defense against microbial pathogens are DNA methylation, histone modification and the activity of non-coding RNAs. There is also growing evidence that opportunistic chronic pathogens, such as Porphyromonas gingivalis, as a microbial host subversion strategy, can epigenetically interfere with the host DNA machinery for successful colonization. Similarly, the novel involvement of small molecule 'danger signals', which are released by stressed or infected cells, at the center of host-pathogen interplay and epigenetics is developing. In this review, we systematically examine the latest knowledge within the field of epigenetics in the context of host-derived danger molecule and purinergic signaling, with a particular focus on host microbial defenses and infection-driven chronic inflammation.
Collapse
Affiliation(s)
- Ana Carolina Morandini
- Department of Biological Sciences, Bauru School of Dentistry - University of São Paulo, Bauru, SP, Brazil Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA 94103, USA Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Carlos F Santos
- Department of Biological Sciences, Bauru School of Dentistry - University of São Paulo, Bauru, SP, Brazil
| | - Özlem Yilmaz
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
95
|
Elsakkar MG, Eissa MM, Hewedy WA, Nassra RM, Elatrebi SF. Sodium valproate, a histone deacetylase inhibitor, with praziquantel ameliorates Schistosoma mansoni-induced liver fibrosis in mice. Life Sci 2016; 162:95-101. [PMID: 27528511 DOI: 10.1016/j.lfs.2016.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/07/2016] [Accepted: 08/10/2016] [Indexed: 11/17/2022]
Abstract
AIMS This study explores the potential antifibrotic effect of sodium valproate (SV), an inhibitor of class I histone deacetylase (HDAC) enzymes, and/or praziquantel (PZQ) on Schistosoma mansoni (S. mansoni)-induced liver fibrosis in mice. MAIN METHODS Male Swiss albino mice were divided into nine groups: group I- normal control (NC); group II- uninfected gum mucilage (GM) treated; group III- uninfected PZQ- treated; group IV- uninfected SV-treated; group V- control S. mansoni infected mice; group VI- infected GM-treated; group VII- infected PZQ-treated; group VIII- infected SV-treated; group IX- infected PZQ+SV treated. All SV administrations were 300mg/kg/day orally and administered for five weeks beginning on the 5th week post infection (WPI). All PZQ administrations were 500mg/kg/day orally and administered for 2 consecutive days beginning on the 7th WPI. Serum transforming growth factor-beta 1 (TGF-β1), tumor necrosis factor-alpha (TNF-α), hepatic hydroxyproline (Hyp) content, and liver function tests (AST and ALT) were determined. Specimens of the hepatic tissues were examined histologically. KEY FINDINGS Treatment of S. mansoni-infected mice with SV significantly decreased the serum levels of ALT, TGF-β1 and TNF-α, and the liver tissue hydroxyproline content compared with the S. mansoni infected untreated groups. Histologically, treatment with SV revealed regression of the granulomatous inflammatory reaction. Combined treatment with PZQ and SV produces more favorable biochemical results, and aborted granulomatous reaction compared with either drug alone. SIGNIFICANCE Sodium valproate is a promising anti-fibrotic agent. It demonstrated an anti-fibrotic effect in early stages of S. mansoni infection through downregulation of profibrogenic cytokines, and collagen deposition.
Collapse
Affiliation(s)
- Mohamed G Elsakkar
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Maha M Eissa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Wafaa A Hewedy
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Rasha M Nassra
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Soha F Elatrebi
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
96
|
Singh N, Johnstone DB, Martin KA, Tempera I, Kaplan MJ, Denny MF. Alterations in nuclear structure promote lupus autoimmunity in a mouse model. Dis Model Mech 2016; 9:885-97. [PMID: 27483354 PMCID: PMC5007980 DOI: 10.1242/dmm.024851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 06/01/2016] [Indexed: 02/06/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by the development of autoantibodies that recognize components of the cell nucleus. The vast majority of lupus research has focused on either the contributions of immune cell dysfunction or the genetics of the disease. Because granulocytes isolated from human SLE patients had alterations in neutrophil nuclear morphology that resembled the Pelger-Huet anomaly, and had prominent mis-splicing of mRNA encoding the nuclear membrane protein lamin B receptor (LBR), consistent with their Pelger-Huet-like nuclear morphology, we used a novel mouse model system to test the hypothesis that a disruption in the structure of the nucleus itself also contributes to the development of lupus autoimmunity. The lupus-prone mouse strain New Zealand White (NZW) was crossed with c57Bl/6 mice harboring a heterozygous autosomal dominant mutation in Lbr (B6.Lbr(ic/+)), and the (NZW×B6.Lbr(ic))F1 offspring were evaluated for induction of lupus autoimmunity. Only female (NZW×B6.Lbr(ic))F1 mice developed lupus autoimmunity, which included splenomegaly, kidney damage and autoantibodies. Kidney damage was accompanied by immune complex deposition, and perivascular and tubule infiltration of mononuclear cells. The titers of anti-chromatin antibodies exceeded those of aged female MRL-Fas(lpr) mice, and were predominantly of the IgG2 subclasses. The anti-nuclear antibody staining profile of female (NZW×B6.Lbr(ic))F1 sera was complex, and consisted of an anti-nuclear membrane reactivity that colocalized with the A-type lamina, in combination with a homogeneous pattern that was related to the recognition of histones with covalent modifications that are associated with gene activation. An anti-neutrophil IgM recognizing calreticulin, but not myeloperoxidase (MPO) or proteinase 3 (PR3), was also identified. Thus, alterations in nuclear structure contribute to lupus autoimmunity when expressed in the context of a lupus-prone genetic background, suggesting a mechanism for the development of lupus autoimmunity in genetically predisposed individuals that is induced by the disruption of nuclear architecture.
Collapse
MESH Headings
- Animals
- Autoantibodies/blood
- Autoantibodies/immunology
- Autoantigens/blood
- Autoantigens/immunology
- Autoimmunity
- Calreticulin/metabolism
- Cell Nucleus/pathology
- Cell Separation
- Crosses, Genetic
- Disease Models, Animal
- Female
- Granulocytes/metabolism
- Granulocytes/pathology
- Histones/metabolism
- Humans
- Immunoglobulin M/immunology
- Kidney/pathology
- Lamin Type A/metabolism
- Lupus Erythematosus, Systemic/blood
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/pathology
- Male
- Mice, Inbred C57BL
- Myeloblastin/metabolism
- Peroxidase/metabolism
- RNA Splicing/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Splenomegaly/pathology
- Transcriptional Activation
- Lamin B Receptor
Collapse
Affiliation(s)
- Namrata Singh
- Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Duncan B Johnstone
- Section of Nephrology, Internal Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Kayla A Martin
- Department of Microbiology/Immunology, Fels Institute for Cancer Research, Temple University, Philadelphia, PA 19140, USA
| | - Italo Tempera
- Department of Microbiology/Immunology, Fels Institute for Cancer Research, Temple University, Philadelphia, PA 19140, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael F Denny
- Section of Rheumatology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| |
Collapse
|
97
|
Fan HC, Lee HS, Chang KP, Lee YY, Lai HC, Hung PL, Lee HF, Chi CS. The Impact of Anti-Epileptic Drugs on Growth and Bone Metabolism. Int J Mol Sci 2016; 17:E1242. [PMID: 27490534 PMCID: PMC5000640 DOI: 10.3390/ijms17081242] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/20/2016] [Accepted: 07/28/2016] [Indexed: 12/13/2022] Open
Abstract
Epilepsy is a common neurological disorder worldwide and anti-epileptic drugs (AEDs) are always the first choice for treatment. However, more than 50% of patients with epilepsy who take AEDs have reported bone abnormalities. Cytochrome P450 (CYP450) isoenzymes are induced by AEDs, especially the classical AEDs, such as benzodiazepines (BZDs), carbamazepine (CBZ), phenytoin (PT), phenobarbital (PB), and valproic acid (VPA). The induction of CYP450 isoenzymes may cause vitamin D deficiency, hypocalcemia, increased fracture risks, and altered bone turnover, leading to impaired bone mineral density (BMD). Newer AEDs, such as levetiracetam (LEV), oxcarbazepine (OXC), lamotrigine (LTG), topiramate (TPM), gabapentin (GP), and vigabatrin (VB) have broader spectra, and are safer and better tolerated than the classical AEDs. The effects of AEDs on bone health are controversial. This review focuses on the impact of AEDs on growth and bone metabolism and emphasizes the need for caution and timely withdrawal of these medications to avoid serious disabilities.
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pediatrics, Tungs' Taichung Metroharbor Hospital, Wuchi, 435 Taichung, Taiwan.
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, 356 Miaoli, Taiwan.
| | - Herng-Shen Lee
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, 813 Kaohsiung, Taiwan.
| | - Kai-Ping Chang
- Department of Pediatrics, Taipei Veterans General Hospital, 112 Taipei, Taiwan.
| | - Yi-Yen Lee
- Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, 112 Taipei, Taiwan.
- Faculty of Medicine, National Yang-Ming University, 112 Taipei, Taiwan.
| | - Hsin-Chuan Lai
- Department of Pediatrics, Tungs' Taichung Metroharbor Hospital, Wuchi, 435 Taichung, Taiwan.
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, 356 Miaoli, Taiwan.
| | - Pi-Lien Hung
- Department of Pediatrics, Kaohsiung Chang Gung Medical Center, 833 Kaohsiung, Taiwan.
| | - Hsiu-Fen Lee
- Department of Pediatrics, Taichung Veterans General Hospital, 407 Taichung, Taiwan.
| | - Ching-Shiang Chi
- Department of Pediatrics, Tungs' Taichung Metroharbor Hospital, Wuchi, 435 Taichung, Taiwan.
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, 356 Miaoli, Taiwan.
| |
Collapse
|
98
|
Pluchino LA, Choudhary S, Wang HCR. Reactive oxygen species-mediated synergistic and preferential induction of cell death and reduction of clonogenic resistance in breast cancer cells by combined cisplatin and FK228. Cancer Lett 2016; 381:124-32. [PMID: 27477899 DOI: 10.1016/j.canlet.2016.07.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 07/25/2016] [Accepted: 07/26/2016] [Indexed: 02/02/2023]
Abstract
Safe and effective combination chemotherapy regimens against breast cancer are lacking. We used our cellular system, consisting of the non-cancerous human breast epithelial MCF10A cell line and its derived tumorigenic, oncogenic H-Ras-expressing, MCF10A-Ras cell line, to investigate the effectiveness of a combination chemotherapy regimen in treating breast cancer cells using two FDA-approved agents, cisplatin and FK228. Cisplatin and FK228 significantly, synergistically, and preferentially induced death and reduced drug resistance of MCF10A-Ras versus MCF10A cells. The ERK-Nox-ROS pathway played a major role in both synergistic cell death induction and GSH-level reduction, which contributed to the synergistic suppression of drug resistance in cells. Enhancement of the Ras-ERK-Nox pathway by combined cisplatin and FK228 significantly increased ROS levels, leading to induction of death, reduction of drug resistance, and induction of DNA damage and oxidation in cancerous MCF10A-Ras cells. Furthermore, synergistic induction of cell death and reduction of drug resistance by combined cisplatin and FK228 in breast cells is independent of their estrogen receptor status. Our study suggests that combined cisplatin and FK228 should be considered in clinical trials as a new regimen for therapeutic control of breast cancers.
Collapse
Affiliation(s)
- Lenora Ann Pluchino
- Anticancer Molecular Oncology Laboratory, Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Shambhunath Choudhary
- Anticancer Molecular Oncology Laboratory, Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Hwa-Chain Robert Wang
- Anticancer Molecular Oncology Laboratory, Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA.
| |
Collapse
|
99
|
Boskovic ZV, Kemp MM, Freedy AM, Viswanathan VS, Pop MS, Fuller JH, Martinez NM, Figueroa Lazú SO, Hong JA, Lewis TA, Calarese D, Love JD, Vetere A, Almo SC, Schreiber SL, Koehler AN. Inhibition of Zinc-Dependent Histone Deacetylases with a Chemically Triggered Electrophile. ACS Chem Biol 2016; 11:1844-51. [PMID: 27064299 DOI: 10.1021/acschembio.6b00012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Unbiased binding assays involving small-molecule microarrays were used to identify compounds that display unique patterns of selectivity among members of the zinc-dependent histone deacetylase family of enzymes. A novel, hydroxyquinoline-containing compound, BRD4354, was shown to preferentially inhibit activity of HDAC5 and HDAC9 in vitro. Inhibition of deacetylase activity appears to be time-dependent and reversible. Mechanistic studies suggest that the compound undergoes zinc-catalyzed decomposition to an ortho-quinone methide, which covalently modifies nucleophilic cysteines within the proteins. The covalent nature of the compound-enzyme interaction has been demonstrated in experiments with biotinylated probe compound and with electrospray ionization-mass spectrometry.
Collapse
Affiliation(s)
- Zarko V. Boskovic
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
- Department
of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Melissa M. Kemp
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Allyson M. Freedy
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
- Department
of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Vasanthi S. Viswanathan
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Marius S. Pop
- Koch
Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Jason H. Fuller
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Nicole M. Martinez
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Samuel O. Figueroa Lazú
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Jiyoung A. Hong
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
- Koch
Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Division
of Hematology/Oncology, Boston Children’s Hospital, Boston, Massachusetts 02116, United States
| | - Timothy A. Lewis
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Daniel Calarese
- Department
of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - James D. Love
- Department
of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Amedeo Vetere
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
| | - Steven C. Almo
- Department
of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461, United States
| | - Stuart L. Schreiber
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
- Department
of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Angela N. Koehler
- Center
for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts 02142, United States
- Koch
Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
100
|
Pro B, Horwitz SM, Prince HM, Foss FM, Sokol L, Greenwood M, Caballero D, Morschhauser F, Wilhelm M, Iyer SP, Shustov AR, Wolfson J, Balser BE, Coiffier B. Romidepsin induces durable responses in patients with relapsed or refractory angioimmunoblastic T-cell lymphoma. Hematol Oncol 2016; 35:914-917. [PMID: 27402335 PMCID: PMC5763404 DOI: 10.1002/hon.2320] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/23/2016] [Accepted: 05/27/2016] [Indexed: 11/24/2022]
Affiliation(s)
- Barbara Pro
- Robert H. Laurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Steven M Horwitz
- Lymphoma Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - H Miles Prince
- Center for Blood Cell Therapies, Peter MacCallum Cancer Center, Melbourne, VIC, Australia
| | | | - Lubomir Sokol
- Malignant Hematology, Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Matthew Greenwood
- Department of Hematology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | | | - Franck Morschhauser
- Hematology, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Martin Wilhelm
- Paracelsus Medical University, Klinikum Nuernberg, Nuernberg, Germany
| | | | - Andrei R Shustov
- Clinical Research Division, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|