251
|
Liu TQ, Sun MJ, Wei MJ. Clinical pathological significance of expression of histone deacetylase 1 and 2 in colorectal adenoma and carcinoma. Shijie Huaren Xiaohua Zazhi 2010; 18:3195-3199. [DOI: 10.11569/wcjd.v18.i30.3195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the clinicopathological significance of expression of histone deacetylase 1 (HDAC1) and HDAC2 in colorectal adenoma and carcinoma.
METHODS: The expression of HDAC1 and HDAC2 in normal colorectal tissue, colorectal adenoma and carcinoma was determined by immunohistochemistry. Immunoreactivity was evaluated semiquantitatively using a staining index (SI). The correlation of the expression of the two epigenetic biomarkers with various clinicalpathological parameters in colorectal adenoma and carcinoma, such as age, tumor size and stage, was analyzed, and survival curves were calculated by the Kaplan-Meier method and the Log-rank test.
RESULTS: The expression of HDAC1 and HDAC2 was significantly lower in normal colorectal tissue than in colorectal adenoma and carcinoma (14.3 ± 9.3 vs 22.4 ± 12.4 and 22.8 ± 8.5; 5.6 ± 3.3 vs 12.3 ± 4.2 and 16.2 ± 9.7, all P < 0.05). The expression of HDAC2 increased in the following order: normal colorectal tissue, colorectal adenoma and carcinoma. The expression of HDAC1 was significantly higher in tumors ≥5 cm than in those < 5 cm (25.1 ± 8.2 vs 20.4 ± 8.5, P < 0.05).
CONCLUSION: Up-regulation of HDAC1 and HDAC2 expression plays a significant role in the development and progression of colorectal carcinoma.
Collapse
|
252
|
Garg D, Henrich S, Salo-Ahen OMH, Myllykallio H, Costi MP, Wade RC. Novel approaches for targeting thymidylate synthase to overcome the resistance and toxicity of anticancer drugs. J Med Chem 2010; 53:6539-49. [PMID: 20527892 DOI: 10.1021/jm901869w] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Divita Garg
- Molecular and Cellular Modeling Group, Heidelberg Institute of Theoretical Studies gGmbH, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
253
|
Upregulated histone deacetylase 1 expression in pancreatic ductal adenocarcinoma and specific siRNA inhibits the growth of cancer cells. Pancreas 2010; 39:994-1001. [PMID: 20467347 DOI: 10.1097/mpa.0b013e3181db0086] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES So far, there are no investigations about the role of histone deacetylase 1 (HDAC1) in tumorigenesis of pancreatic ductal adenocarcinoma. This study was designed to elucidate the roles and mechanisms of HDAC1 in tumorigenesis of pancreatic ductal adenocarcinoma. METHODS Real-time reverse transcription-polymerase chain reaction and immunohistochemistry techniques were adopted to detect the expression of HDAC1 in human pancreatic ductal adenocarcinoma tissues and paired paracancerous tissues. The roles of HDAC1 in human pancreatic cell line PaTu8988 were investigated using siRNA. RESULTS Histone deacetylase 1 mRNA in pancreatic cancer tissues were significantly higher than in paracancerous tissues (P < 0.05). Immunohistochemistry showed that the indices of HDAC1 in pancreatic cancer tissues and paracancerous tissues were 56.4% (SD, 23.1%) and 6.7% (SD, 5.0%), respectively (P < 0.001). Knockdown of HDAC1 can generate a remarkable defect in proliferation and also can significantly induce apoptosis and S-phase arrest in PaTu8988 cells (P < 0.05). The Bcl-2 mRNA expression was significantly downregulated, whereas the p21 and Bax mRNA expression were significantly upregulated. CONCLUSIONS The HDAC1 overexpression might play an important role in tumorigenesis of pancreatic cancer. Our data support the development of selective inhibitors targeting HDAC1 for the treatment of pancreatic ductal adenocarcinoma. Histone deacetylase 1 could be a new gene therapy target in pancreatic ductal adenocarcinoma.
Collapse
|
254
|
Zhang L, Fang H, Xu W. Strategies in developing promising histone deacetylase inhibitors. Med Res Rev 2010; 30:585-602. [PMID: 19634125 DOI: 10.1002/med.20169] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDACs) are a family of enzymes that have been of interest in drug discovery for more than 30 years. Inhibitors of HDACs are potential therapeutics for various diseases, such as neurodegenerative diseases, inflammation, viral infection, and especially cancer. Most HDAC inhibitors (HDACi) are designed for cancer therapy. In 2006, suberoylanilide hydroxamic acid was approved by the US Food and Drug Administration for once-daily oral treatment of advanced cutaneous T-cell lymphoma. In the meantime, there have been aggressive efforts to bring HDACi to the market for every major tumor type, either as a single therapy or in combination, and a number of compounds are currently undergoing clinical trials. Multiple strategies have been applied to the rational design of drugs targeting HDACs by taking advantage of the new developments in proteomics, chemogenomics, cheminformatics, and computational chemistry/biology. Herein, we review the current methods successfully used in developing novel HDACi.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Medicinal Chemistry, School of Pharmacy, ShanDong University, No. 44, West Culture Road, Ji'nan, ShanDong, P.R. China
| | | | | |
Collapse
|
255
|
Chia K, Beamish H, Jafferi K, Gabrielli B. The histone deacetylase inhibitor MGCD0103 has both deacetylase and microtubule inhibitory activity. Mol Pharmacol 2010; 78:436-43. [PMID: 20538840 DOI: 10.1124/mol.110.065169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Histone deacetylase inhibitors (HDACis) are currently in trial or are in clinical use for the treatment of a number of tumor types. The clinical efficacy of HDACis can be partly attributed to the modulation of the cell cycle by the HDACis. Here, we have examined the effects of N-(2-aminophenyl)-4-((4-pyridin-3-ylpyrimidin-2-ylamino)methyl)benzamide (MGCD0103), a class I-selective histone deacetylase inhibitor, on the cell cycle and cell killing. Surprisingly, MGCD0103 treatment failed to initiate a G(1)-phase arrest but caused marked accumulation of cells in G(2)/M at 6 and 12 h after treatment and was cytotoxic 24 h after treatment. These cell cycle effects were considerably distinct from the effects of suberic bishydroxamic acid, a representative of the pan-isoform HDACi used in this study. MGCD0103 shared the ability of the pan-isoform HDACi to trigger defective mitosis and promote mitotic slippage. Likewise, it also specifically targeted tumor cells and was nontoxic to normal nontransformed cells. However, MGDC0103 also seemed to disrupt normal microtubule spindle formation, whereas HDACis generally have only a minor effect on spindle formation. The effect of MGCD0103 on spindle formation was shown to be a consequence of microtubule destabilization. This is the first example of an HDACi with microtubule destabilizing activity, and the combined effects of this drug have advantages for its therapeutic use.
Collapse
Affiliation(s)
- Keeming Chia
- University of Queensland Diamantina Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | | | | | | |
Collapse
|
256
|
Altmann A, Eisenhut M, Bauder-Wüst U, Markert A, Askoxylakis V, Hess-Stumpp H, Haberkorn U. Therapy of thyroid carcinoma with the histone deacetylase inhibitor MS-275. Eur J Nucl Med Mol Imaging 2010; 37:2286-97. [DOI: 10.1007/s00259-010-1573-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 07/15/2010] [Indexed: 01/01/2023]
|
257
|
Kiyokawa S, Hirata Y, Nagaoka Y, Shibano M, Taniguchi M, Yasuda M, Baba K, Uesato S. New orally bioavailable 2-aminobenzamide-type histone deacetylase inhibitor possessing a (2-hydroxyethyl)(4-(thiophen-2-yl)benzyl)amino group. Bioorg Med Chem 2010; 18:3925-33. [PMID: 20452226 DOI: 10.1016/j.bmc.2010.04.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 04/12/2010] [Accepted: 04/13/2010] [Indexed: 11/26/2022]
Abstract
New 2-aminobenzamide-type histone deacetylase (HDAC) inhibitors were synthesized. They feature a sulfur-containing bicyclic arylmethyl moiety-a surface recognition domain introduced to increase in cellular uptake-and a substituted tert-amino group which affects physicochemical properties such as aqueous solubility. Compound 22 with a (2-hydroxyethyl)(4-(thiophen-2-yl)benzyl)amino group reduced the volume of human colon cancer HCT116 xenografts in nude mice to T/C 67% by oral administration at 45mg/kg, which was comparable to the rate (T/C 62%) for a positive control, MS-275. Western blot analyses as well as cell cycle and TUNEL assays by flow cytometry suggested that the two compounds inhibited the growth of cancer cells via similar mechanisms.
Collapse
Affiliation(s)
- Shingo Kiyokawa
- Department of Life Science and Biotechnology, Kansai University, Suita, Osaka 564-8680, Japan
| | | | | | | | | | | | | | | |
Collapse
|
258
|
Wang H, Lim ZY, Zhou Y, Ng M, Lu T, Lee K, Sangthongpitag K, Goh KC, Wang X, Wu X, Khng HH, Goh SK, Ong WC, Bonday Z, Sun ET. Acylurea connected straight chain hydroxamates as novel histone deacetylase inhibitors: Synthesis, SAR, and in vivo antitumor activity. Bioorg Med Chem Lett 2010; 20:3314-21. [DOI: 10.1016/j.bmcl.2010.04.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 04/01/2010] [Accepted: 04/10/2010] [Indexed: 10/19/2022]
|
259
|
Lee CK, Wang S, Huang X, Ryder J, Liu B. HDAC inhibition synergistically enhances alkylator-induced DNA damage responses and apoptosis in multiple myeloma cells. Cancer Lett 2010; 296:233-40. [PMID: 20447761 DOI: 10.1016/j.canlet.2010.04.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2010] [Revised: 04/07/2010] [Accepted: 04/08/2010] [Indexed: 01/12/2023]
Abstract
Histone deacetylase (HDAC) inhibitors induce chromatin destabilization. We sought to determine whether HDAC inhibition may amplify alkylator-induced mitotic cell death in multiple myeloma (MM) cells. The combination of SNDX-275, a class I HDAC inhibitor, with melphalan, showed a powerful synergism on growth inhibition with the combination index ranged from 0.27 to 0.75 in MM1.S and RPMI8226 cells. Their combinations as compared with either agent alone promoted much more caspase-dependent apoptosis. Flow cytometry analysis showed that SNDX-275 had minimal effects on cell cycle progression of MM1.S cells, but clearly increased the percentage of S phase in RPMI8226 cells associated with an upregulation in p21(waf1) and a reduction in cyclin D1 and E2F1. Melphalan alone significantly arrested both MM1.S and RPMI8226 cells at S phase and enhanced expression of p53 and p21(waf1). Furthermore, studies on DNA damage response revealed that phospho-histone H2A.X (gammaH2A.X), a hall marker of DNA double strand break, along with phosphorylated CHK1 (P-CHK1) and CHK2 (P-CHK2) was dramatically induced by SNDX-275 or melphalan. The increase in gammaH2A.X and P-CHK1 was considerably higher on combination than either agent alone. These molecular changes correlated well with the significant increase in mitotic catastrophe. Our data indicate that SNDX-275 synergistically enhances melphalan-induced apoptosis in MM cells via intensification of DNA damage, suggesting that SNDX-275 in combination with melphalan may be a novel therapeutic strategy for MM.
Collapse
Affiliation(s)
- Choon-Kee Lee
- Department of Medicine, University of Colorado Denver School of Medicine, Aurora, 80045, USA.
| | | | | | | | | |
Collapse
|
260
|
Identification of type-specific anticancer histone deacetylase inhibitors: road to success. Cancer Chemother Pharmacol 2010; 66:625-33. [DOI: 10.1007/s00280-010-1324-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 04/01/2010] [Indexed: 10/19/2022]
|
261
|
Huang WJ, Chen CC, Chao SW, Lee SS, Hsu FL, Lu YL, Hung MF, Chang CI. Synthesis ofN-Hydroxycinnamides Capped with a Naturally Occurring Moiety as Inhibitors of Histone Deacetylase. ChemMedChem 2010; 5:598-607. [DOI: 10.1002/cmdc.200900494] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
262
|
Bressi JC, Jennings AJ, Skene R, Wu Y, Melkus R, De Jong R, O'Connell S, Grimshaw CE, Navre M, Gangloff AR. Exploration of the HDAC2 foot pocket: Synthesis and SAR of substituted N-(2-aminophenyl)benzamides. Bioorg Med Chem Lett 2010; 20:3142-5. [PMID: 20392638 DOI: 10.1016/j.bmcl.2010.03.091] [Citation(s) in RCA: 202] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 03/25/2010] [Accepted: 03/26/2010] [Indexed: 11/26/2022]
Abstract
A series of N-(2-amino-5-substituted phenyl)benzamides (3-21) were designed, synthesized and evaluated for their inhibition of HDAC2 and their cytotoxicity in HCT116 cancer cells. Multiple compounds from this series demonstrated time-dependent binding kinetics that is rationalized using a co-complex crystal structure of HDAC2 and N-(4-aminobiphenyl-3-yl)benzamide (6).
Collapse
|
263
|
Desai D, Salli U, Vrana KE, Amin S. SelSA, selenium analogs of SAHA as potent histone deacetylase inhibitors. Bioorg Med Chem Lett 2010; 20:2044-7. [PMID: 20167479 PMCID: PMC2892848 DOI: 10.1016/j.bmcl.2009.07.068] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Revised: 07/09/2009] [Accepted: 07/14/2009] [Indexed: 10/20/2022]
Abstract
Cancer treatment and therapy has moved from conventional chemotherapeutics to more mechanism-based targeted approach. Disturbances in the balance of histone acetyltransferase (HAT) and deacetylase (HDAC) leads to a change in cell morphology, cell cycle, differentiation, and carcinogenesis. In particular, HDAC plays an important role in carcinogenesis and therefore it has been a target for cancer therapy. Structurally diverse group of HDAC inhibitors are known. The broadest class of HDAC inhibitor belongs to hydroxamic acid derivatives that have been shown to inhibit both class I and II HDACs. Suberoylanilide hydroxamic acid (SAHA) and Trichostatin A (TSA), which chelate the zinc ions, fall into this group. In particular, SAHA, second generation HDAC inhibitor, is in several cancer clinical trials including solid tumors and hematological malignancy, advanced refractory leukemia, metastatic head and neck cancers, and advanced cancers. To our knowledge, selenium-containing HDAC inhibitors are not reported in the literature. In order to find novel HDAC inhibitors, two selenium based-compounds modeled after SAHA were synthesized. We have compared two selenium-containing compounds; namely, SelSA-1 and SelSA-2 for their inhibitory HDAC activities against SAHA. Both, SelSA-1 and SelSA-2 were potent HDAC inhibitors; SelSA-2 having IC50 values of 8.9 nM whereas SAHA showed HDAC IC(50) values of 196 nM. These results provided novel selenium-containing potent HDAC inhibitors.
Collapse
Affiliation(s)
- Dhimant Desai
- Department of Pharmacology, Pennsylvania State Hershey College of Medicine, Hershey, PA 17033, USA.
| | | | | | | |
Collapse
|
264
|
Pontiki E, Hadjipavlou-Litina D. Histone deacetylase inhibitors (HDACIs). Structure--activity relationships: history and new QSAR perspectives. Med Res Rev 2010; 32:1-165. [PMID: 20162725 DOI: 10.1002/med.20200] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Histone deacetylase (HDAC) inhibition is a recent, clinically validated therapeutic strategy for cancer treatment. HDAC inhibitors (HDACIs) block angiogenesis, arrest cell growth, and lead to differentiation and apoptosis in tumor cells. In this article, a survey of published quantitative structure-activity relationships (QSARs) studies are presented and discussed in the hope of identifying the structural determinants for anticancer activity. Secondly a two-dimensional QSAR study was carried out on biological results derived from various types of HDACIs and from different assays using the C-QSAR program of Biobyte. The QSAR analysis presented here is an attempt to organize the knowledge on the HDACIs with the purpose of designing new chemical entities with enhanced inhibitory potencies and to study the mechanism of action of the compounds. This study revealed that lipophilicity is one of the most important determinants of activity. Additionally, steric factors such as the overall molar refractivity (CMR), molar volume (MgVol), the substituent's molar refractivity (MR) (linear or parabola), or the sterimol parameters B(1) and L are important. Electronic parameters indicated as σ(p), are found to be present only in one case.
Collapse
Affiliation(s)
- Eleni Pontiki
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotelian University of Thessaloniki, Thessaloniki 54124, Greece.
| | | |
Collapse
|
265
|
Preclinical studies of chemotherapy using histone deacetylase inhibitors in endometrial cancer. Obstet Gynecol Int 2010; 2010:923824. [PMID: 20169171 PMCID: PMC2821626 DOI: 10.1155/2010/923824] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 11/23/2009] [Accepted: 01/14/2010] [Indexed: 11/29/2022] Open
Abstract
Because epigenetic alterations are believed to be involved in the repression of tumor suppressor genes and promotion of tumorigenesis in endometrial cancers, novel compounds endowed with a histone deacetylase (HDAC) inhibitory activity are an attractive therapeutic approach. In this review, we discuss the biologic and therapeutic effects of HDAC inhibitors (HDACIs) in treating endometrial cancer. HDACIs were able to mediate inhibition of cell growth, cell cycle arrest, apoptosis, and the expression of genes related to the malignant phenotype in a variety of endometrial cancer cell lines. Furthermore, HDACIs were able to induce the accumulation of acetylated histones in the chromatin of the p21WAF1 gene in human endometrial carcinoma cells. In xenograft models, some HDACIs have demonstrated antitumor activity with only few side effects. In this review, we discuss the biologic and therapeutic effects of HDACIs in treating endometrial cancer, with a special focus on preclinical studies.
Collapse
|
266
|
WATANABE M, CHEN IYJ, ISHIKURA Y, NAKAGAWA T, MOCHIZUKI M, NISHIMURA R, SASAKI N, SUGANO S. Tumor cell growth inhibition and cell differentiation analysis in a canine mammary tumor cell line (MCM-B2) treated with four chemical reagents. J Vet Med Sci 2010; 71:1413-7. [PMID: 19959889 DOI: 10.1292/jvms.001413] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In this study, we examined the effects of four chemical reagents, 5 azacytidine (5azaC), all-trans-retinoic acid (ATRA), Phorbol 12-myristate 13-acetate (TPA) and trichostatin A (TSA), on an MCM-B2 canine mammary tumor cell line. Growth of the MCM-B2 cells was inhibited by addition of any of these four chemical reagents in a dose-dependent manner. TPA-treated cells became blast-cell-like and had high expressions of cyclin D, cyclin B and PCNA. Both ATRA-treated and 5azaC-treated cells showed decreased expression of cell cycle related molecules and increased expressions of the mammary epithelial marker cytokeratin 18 and underwent morphological changes. ATRA-treated cells were converted from the originally spindle-shaped cells into cubic-shaped cells, and 5azaC-treated cells became fibroblast-like. In the TSA-treated cells, the cytoplasm was enlarged, increasing the cytoplasm/nucleus ratio, and the expressions of cell cycle-promoting molecules (cyclin D and PCNA), as well as cell cycle-inhibiting factors (p21(WAF1) and p27(kip1)), were increased. These results demonstrated the growth-inhibiting, differentiation-inducing and antitumor effects of each of these four chemicals on the unique phenotype of canine mammary tumors.
Collapse
Affiliation(s)
- Manabu WATANABE
- Laboratory of Functional Genomics, Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Minato-ku, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
267
|
Kim IA, Kim IH, Kim HJ, Chie EK, Kim JS. HDAC inhibitor-mediated radiosensitization in human carcinoma cells: a general phenomenon? JOURNAL OF RADIATION RESEARCH 2010; 51:257-263. [PMID: 20505264 DOI: 10.1269/jrr.09115] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Histone deacetylase inhibitors (HDIs) have attracted considerable attention for anticancer therapy strategy, including radiosensitization. Regarding a potential application of HDI as a radiosensitizer in the treatment of solid tumors, an important question is whether treatment efficacy would be influenced by intrinsic differences between cancer cells, such as different histologic origin and status of ATM or p53. First we have observed the in vitro radiosensitization by Trichostatin A (TSA) on the broad spectrum of human tumor cell lines having different histologic origin such as HCT116 adenocarcinoma of colon, A549 adenocarcinoma of lung, HN-3 squamous cell carcinoma of head/neck, and HeLa squamous carcinoma of uterine cervix, using clonogenic assay. Next, we have systematically assessed the radiosensitization on the cell lines having different ATM or p53 status. We found that pretreatment of HDI consistently resulted in radiosensitization of all cell lines tested, though the sensitizer enhancement ratio of individual cell lines was variable. We also observed that TSA-mediated radiosensitization was clearly influenced by p53 and ATM status of cells tested. The data presented here indicate that HDI enhances the radiation induced cell killing in the various cancer cells having intrinsic differences and may serve as a general strategy for enhancing tumor cell radiosensitivity. These results have potential implications for the clinical utility of HDI in increasing the anticancer efficacy of radiation.
Collapse
Affiliation(s)
- In Ah Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
268
|
Mwakwari SC, Patil V, Guerrant W, Oyelere AK. Macrocyclic histone deacetylase inhibitors. Curr Top Med Chem 2010; 10:1423-40. [PMID: 20536416 PMCID: PMC3144151 DOI: 10.2174/156802610792232079] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2010] [Accepted: 05/08/2010] [Indexed: 01/12/2023]
Abstract
Histone deacetylase inhibitors (HDACi) are an emerging class of novel anti-cancer drugs that cause growth arrest, differentiation, and apoptosis of tumor cells. In addition, they have shown promise as anti-parasitic, anti-neurodegenerative, anti-rheumatologic and immunosuppressant agents. To date, several structurally distinct small molecule HDACi have been reported including aryl hydroxamates, benzamides, short-chain fatty acids, electrophilic ketones, and macrocyclic peptides. Macrocyclic HDACi possess the most complex cap-groups which interact with HDAC enzyme's outer rim and have demonstrated excellent HDAC inhibition potency and isoform selectivity. This review focuses on the recent progress and current state of macrocyclic HDACi.
Collapse
Affiliation(s)
- Sandra C. Mwakwari
- School of Chemistry and Biochemistry, Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400
| | - Vishal Patil
- School of Chemistry and Biochemistry, Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400
| | - William Guerrant
- School of Chemistry and Biochemistry, Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400
| | - Adegboyega K. Oyelere
- School of Chemistry and Biochemistry, Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400
| |
Collapse
|
269
|
Hooker JM, Kim SW, Alexoff D, Xu Y, Shea C, Reid A, Volkow N, Fowler JS. Histone deacetylase inhibitor, MS-275, exhibits poor brain penetration: PK studies of [C]MS-275 using Positron Emission Tomography. ACS Chem Neurosci 2010; 1:65-73. [PMID: 20657706 PMCID: PMC2908422 DOI: 10.1021/cn9000268] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 10/14/2009] [Indexed: 01/05/2023] Open
Abstract
MS-275 (Entinostat) is a histone deacetylase (HDAC) inhibitor currently in clinical trials for the treatment of several types of cancer. Recent reports have noted that MS-275 can cross the blood brain barrier (BBB) and cause region specific changes in rodent brain histone acetylation. To characterize the pharmacokinetics and distribution of MS-275 in the brain using positron emission tomography (PET), we labeled the carbamate carbon of MS-275 with carbon-11. Using PET, we determined that [(11)C]MS-275 has low uptake in brain tissue when administered intravenously to non-human primates. In rodent studies, we observed that pharmacokinetics and brain accumulation of [(11)C]MS-275 were not changed by the co-administration of large doses of unlabeled MS-275. These results, which both highlight the poor brain penetration of MS-275, clearly suggest its limitation as a therapeutic agent for the central nervous system (CNS). Moreover, our study demonstrates the effectiveness of PET at providing brain pharmacokinetic data for HDAC inhibitors. These data are important not only for the development of new compounds for peripheral cancer treatment (where CNS exclusion is often advantageous), but also for the treatment of neurological disorders (where CNS penetration is critical).
Collapse
Affiliation(s)
- Jacob M. Hooker
- Medical Department, Brookhaven National Laboratory, Upton, New York 11973
| | - Sung Won Kim
- Medical Department, Brookhaven National Laboratory, Upton, New York 11973
- National Institute on Alcohol Abuse and Alcoholism, Rockville, Maryland 20892
| | - David Alexoff
- Medical Department, Brookhaven National Laboratory, Upton, New York 11973
| | - Youwen Xu
- Medical Department, Brookhaven National Laboratory, Upton, New York 11973
| | - Colleen Shea
- Medical Department, Brookhaven National Laboratory, Upton, New York 11973
| | - Alicia Reid
- Medical Department, Brookhaven National Laboratory, Upton, New York 11973
- School of Science, Health and Technology, Medgar Evers College, Brooklyn, New York 11225
| | - Nora Volkow
- Medical Department, Brookhaven National Laboratory, Upton, New York 11973
- National Institute on Alcohol Abuse and Alcoholism, Rockville, Maryland 20892
- National Institute on Drug Abuse, Rockville, Maryland 20892
| | - Joanna S. Fowler
- Medical Department, Brookhaven National Laboratory, Upton, New York 11973
| |
Collapse
|
270
|
Ohtsuki A, Kimura MT, Minoshima M, Suzuki T, Ikeda M, Bando T, Nagase H, Shinohara KI, Sugiyama H. Synthesis and properties of PI polyamide–SAHA conjugate. Tetrahedron Lett 2009. [DOI: 10.1016/j.tetlet.2009.10.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
271
|
Bergamaschi A, David A, Le Rouzic E, Nisole S, Barré-Sinoussi F, Pancino G. The CDK inhibitor p21Cip1/WAF1 is induced by FcgammaR activation and restricts the replication of human immunodeficiency virus type 1 and related primate lentiviruses in human macrophages. J Virol 2009; 83:12253-65. [PMID: 19759136 PMCID: PMC2786717 DOI: 10.1128/jvi.01395-09] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 09/10/2009] [Indexed: 12/26/2022] Open
Abstract
Macrophages are major targets of human immunodeficiency virus type 1 (HIV-1). We have previously shown that aggregation of activating immunoglobulin G Fc receptors (FcgammaR) by immune complexes inhibits reverse transcript accumulation and integration of HIV-1 and related lentiviruses in monocyte-derived macrophages. Here, we show that FcgammaR-mediated restriction of HIV-1 is not due to enhanced degradation of incoming viral proteins or cDNA and is associated to the induction of the cyclin-dependent kinase inhibitor p21(Cip1/WAF1) (p21). Small interfering RNA-mediated p21 knockdown rescued viral replication in FcgammaR-activated macrophages and enhanced HIV-1 infection in unstimulated macrophages by increasing reverse transcript and integrated DNA levels. p21 induction by other stimuli, such as phorbol myristate acetate and the histone deacetylase inhibitor MS-275, was also associated with preintegrative blocks of HIV-1 replication in macrophages. Binding of p21 to reverse transcription/preintegration complex-associated HIV-1 proteins was not detected in yeast two-hybrid, pulldown, or coimmunoprecipitation assays, suggesting that p21 may affect viral replication independently of a specific interaction with an HIV-1 component. Consistently, p21 silencing rescued viral replication from the FcgammaR-mediated restriction also in simian immunodeficiency virus SIV(mac)- and HIV-2-infected macrophages. Our results point to a role of p21 as an inhibitory factor of lentiviral infection in macrophages and to its implication in FcgammaR-mediated restriction.
Collapse
Affiliation(s)
- Anna Bergamaschi
- Institut Pasteur, Unité de Régulation des Infections Rétrovirales, Paris, France, Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Département des Maladies Infectieuses, Paris, France, INSERM, U567, 27 Rue du Faubourg St. Jacques, 75014 Paris, France
| | - Annie David
- Institut Pasteur, Unité de Régulation des Infections Rétrovirales, Paris, France, Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Département des Maladies Infectieuses, Paris, France, INSERM, U567, 27 Rue du Faubourg St. Jacques, 75014 Paris, France
| | - Erwann Le Rouzic
- Institut Pasteur, Unité de Régulation des Infections Rétrovirales, Paris, France, Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Département des Maladies Infectieuses, Paris, France, INSERM, U567, 27 Rue du Faubourg St. Jacques, 75014 Paris, France
| | - Sébastien Nisole
- Institut Pasteur, Unité de Régulation des Infections Rétrovirales, Paris, France, Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Département des Maladies Infectieuses, Paris, France, INSERM, U567, 27 Rue du Faubourg St. Jacques, 75014 Paris, France
| | - Françoise Barré-Sinoussi
- Institut Pasteur, Unité de Régulation des Infections Rétrovirales, Paris, France, Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Département des Maladies Infectieuses, Paris, France, INSERM, U567, 27 Rue du Faubourg St. Jacques, 75014 Paris, France
| | - Gianfranco Pancino
- Institut Pasteur, Unité de Régulation des Infections Rétrovirales, Paris, France, Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Département des Maladies Infectieuses, Paris, France, INSERM, U567, 27 Rue du Faubourg St. Jacques, 75014 Paris, France
| |
Collapse
|
272
|
Schneider G, Krämer OH, Fritsche P, Schüler S, Schmid RM, Saur D. Targeting histone deacetylases in pancreatic ductal adenocarcinoma. J Cell Mol Med 2009; 14:1255-63. [PMID: 19929947 PMCID: PMC3828843 DOI: 10.1111/j.1582-4934.2009.00974.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a dismal disease with a median survival below 6 months and a 5-year survival rate below 1%. Effective therapies for locally advanced or metastatic tumours are missing and curatively resected patients relapse in over 80% of the cases. Although histone deacetylases (HDACs) are involved in the control of proliferation, apoptosis, differentiation, migration and angiogenesis of cancer cells, knowledge about the expression patterns and functions of individual HDAC isoenzymes in pancreatic cancer is sparse. This review summarizes the roles of HDACs as novel therapeutic targets and the molecular mode of action of HDAC-inhibitors (HDACI) in PDACs. Success of HDACI in clinical settings will depend on an increased knowledge of HDAC functions as well as on a better understanding of the mode of action of HDACI. Pre-clinical experimental data that constitute the basis for rational therapeutic strategies to treat PDAC are described here. Translating these rational-based therapies into the clinic will finally increase our chance to establish an effective HDACI-containing combination therapy effective against PDAC.
Collapse
Affiliation(s)
- Günter Schneider
- Technische Universität München, Klinikum rechts der Isar, II. Medizinische Klinik, München, Germany.
| | | | | | | | | | | |
Collapse
|
273
|
Abstract
For the first 30 years since its discovery, reversible protein acetylation has been studied and understood almost exclusively in the context of histone modification and gene transcription. With the discovery of non-histone acetylated proteins and acetylation-modifying enzymes in cellular compartments outside the nucleus, the regulatory potential of reversible acetylation has slowly been recognized in the last decade. However, the scope of protein acetylation involvement in complex biological processes remains uncertain. The recent development of new technology has enabled, for the first time, the identification and quantification of the acetylome, acetylation events at the whole-proteome level. These efforts have uncovered a stunning complexity of the acetylome that potentially rivals that of the phosphoproteome. The remarkably ubiquitous and conserved nature of protein acetylation revealed by these new studies suggests the regulatory power of this dynamic modification. The establishment of comprehensive acetylomes will change the landscape of protein acetylation, where an exciting research frontier awaits.
Collapse
Affiliation(s)
- Kristi L Norris
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | | | | |
Collapse
|
274
|
Histone deacetylase inhibitor prodrugs in nanoparticle vector enhanced gene expression in human cancer cells. Eur J Med Chem 2009; 44:4603-10. [DOI: 10.1016/j.ejmech.2009.06.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Revised: 06/15/2009] [Accepted: 06/28/2009] [Indexed: 11/30/2022]
|
275
|
Li Q, Bartlett DL, Gorry MC, O'Malley ME, Guo ZS. Three epigenetic drugs up-regulate homeobox gene Rhox5 in cancer cells through overlapping and distinct molecular mechanisms. Mol Pharmacol 2009; 76:1072-81. [PMID: 19679824 DOI: 10.1124/mol.109.056291] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Epigenetic therapy of cancer using inhibitors of DNA methyltransferases (DNMT) or/and histone deacetylases (HDACs) has shown promising results in preclinical models and is being investigated in clinical trials. Homeodomain proteins play important roles in normal development and carcinogenesis. In this study, we demonstrated for the first time that an epigenetic drug could up-regulate homeobox genes in the reproductive homeobox genes on chromosome X (Rhox) family, including murine Rhox5, Rhox6, and Rhox9 and human RhoxF1 and RhoxF2 in breast, colon, and other types of cancer cells. We examined the molecular mechanisms underlining selective induction of Rhox5 in cancer cells by three epigenetic drugs: 5-aza-2'-deoxycytidine (DAC; decitabine), arsenic trioxide (ATO), and MS-275 [entinostat; N-(2-aminophenyl)-4-[N-(pyridine-3-ylmethoxy-carbonyl)aminomethyl]benzamide]. DAC induced Rhox5 mRNA expression from both distal promoter (Pd) and proximal promoter, whereas MS-275 and ATO induced gene expression from the Pd only. DAC and ATO inhibited both DNMT1 and DNMT3B protein expression, whereas MS-275 significantly reduced DNMT3B protein. In contrast to DAC, neither MS-275 nor ATO induced DNA demethylation on the Pd region. All three drugs led to enhanced acetylation of histones H3 and H4 at the promoter region. The occupancy of the activating histone mark dimethylated lysine 4 of H3 at Pd was enhanced by DAC and MS-275 but not ATO. Because they modulate gene expression with different potencies through shared and distinct epigenetic mechanisms, these epigenetic drugs may possess great potential in different applications for epigenetic therapy of cancer and other diseases.
Collapse
Affiliation(s)
- Qiang Li
- University of Pittsburgh Cancer Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
276
|
Huang X, Gao L, Wang S, Lee CK, Ordentlich P, Liu B. HDAC inhibitor SNDX-275 induces apoptosis in erbB2-overexpressing breast cancer cells via down-regulation of erbB3 expression. Cancer Res 2009; 69:8403-11. [PMID: 19826038 DOI: 10.1158/0008-5472.can-09-2146] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Breast cancer is a highly heterogeneous disease with distinct histologic subtypes. Targeted therapies such as endocrine therapy and growth factor receptor inhibitors have had a significant impact on the treatment of metastatic breast cancer patients. Unfortunately, resistance to these agents eventually occurs, and currently represents a significant clinical problem in the management of breast cancers. Inhibitors of histone deacetylases (HDACi) exhibit anticancer activity in a variety of tumor cell models and have been shown to target mechanisms of resistance to a number of targeted agents. It is unclear, however, if there are specific breast cancer subtypes for which an HDACi may be more or less effective. Here, we report that the class I isoform-selective HDACi entinostat (SNDX-275) preferentially inhibits cell proliferation/survival and inactivates downstream signaling in erbB2-overexpressing compared with basal breast cancer cells. SNDX-275 reduces the levels of both erbB2 and erbB3, as well as significantly decreases P-erbB2, P-erbB3, P-Akt, and P-MAPK in erbB2-overexpressing cells. Additionally, SNDX-275 promotes apoptosis and induces cell cycle arrest predominantly at G(1) phase in erbB2-overexpressing cells, whereas SNDX-275 mainly induces G(2)-M arrest in basal breast cancer cells. The cellular bias of SNDX-275 is shown to be related partly to the levels of erbB3 expression that directly impact the ability of SNDX-275 to inhibit proliferation/survival of the erbB2-overexpressing breast cancer cells. These findings show that SNDX-275 may be developed as a novel therapeutic agent to treat breast cancers with coexpression of both erbB2 and erbB3.
Collapse
Affiliation(s)
- Xiaoping Huang
- Department of Pathology, University of Colorado Denver School of Medicine, Aurora, Colorado 80045, USA
| | | | | | | | | | | |
Collapse
|
277
|
The synergy of panobinostat plus doxorubicin in acute myeloid leukemia suggests a role for HDAC inhibitors in the control of DNA repair. Leukemia 2009; 23:2265-74. [PMID: 19812608 DOI: 10.1038/leu.2009.182] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute myeloid leukemia (AML) is a clonal disorder characterized by the accumulation of myeloid blasts in the bone marrow. Here, we report the effects of the novel histone deacetylase inhibitor panobinostat (LBH589) in combination with doxorubicin on AML cells. Panobinostat exhibited potent anti-AML activity in all AML cell lines tested and in primary AML cells from patients (IC(50)<20 nM). In addition, panobinostat potentiated the action of several standard-of-care anti-AML compounds, particularly, doxorubicin. The molecular effects induced by panobinostat and doxorubicin treatment were investigated by analyzing gene expression, cell cycle, apoptosis and signaling pathways. Analyses of gene expression profiles identified 588 genes whose expression was exclusively affected by the combination of panobinostat and doxorubicin. The combination induced AML cell death by an increase in the mitochondrial outer membrane permeability and release of cytochrome c from the mitochondria, resulting in caspase-dependent apoptosis and accompanied by the upregulation of Bax, Bak and, particularly, Bad. The drug combination provoked a strong activation of a DNA damage response, indicating that this combination may trigger cell death by a mechanism that induced DNA double-strand breaks. These data indicate that the combination of panobinostat and doxorubicin may be an effective therapy for the treatment of AML.
Collapse
|
278
|
Vire B, de Walque S, Restouin A, Olive D, Van Lint C, Collette Y. Anti-leukemia activity of MS-275 histone deacetylase inhibitor implicates 4-1BBL/4-1BB immunomodulatory functions. PLoS One 2009; 4:e7085. [PMID: 19759901 PMCID: PMC2738963 DOI: 10.1371/journal.pone.0007085] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Accepted: 08/10/2009] [Indexed: 11/18/2022] Open
Abstract
Histone deacetylase inhibitors (HDACi) have demonstrated promising therapeutic potential in clinical trials for hematological malignancies. HDACi, such as SAHA/Vorinostat, Trichostatin A, and MS-275 were found to induce apoptosis of leukemic blasts through activation of the death receptor pathway and transcriptional induction of the Tumor Necrosis Factor (TNF)-related pro-apoptotic family members, TRAIL and FasL. The impact of HDACi on TNF-related costimulatory molecules such as 4-1BB ligand (4-1BBL/TNFSF9) is however not known. Following exposure to SAHA/Vorinostat, Trichostatin A, and MS-275, transcript levels were determined by real time PCR in Jurkat, Raji and U937 cells. Treatment with HDACi up-regulated TNFSF9 gene expression in the three leukemia cell lines, yet to different extend and with distinct kinetics, which did not require de novo protein synthesis and was not associated with DNAse I hypersensitive chromatin remodeling. Transcriptional activity of TNFSF9 promoter-luciferase constructs was induced up to 12 fold by HDACi, and implication of Sp1/Sp3 transcription factors binding to functional GC-box elements was evidenced by reporter gene assays, site-directed mutagenesis, and electrophoretic mobility shift assays. Functionality of modulated target genes was assessed in allogeneic mixed leukocyte reaction experiments. MS-275- and to a lesser extent Trichostatin A- and SAHA-treated Raji cells significantly up regulated T lymphocytes proliferation which was reduced by about 50% by a 4-1BB blocking recombinant protein, while MS-275- but neither Trichostatin A- nor SAHA-treated cells up-regulated IFNγ secretion by T lymphocytes. Our results identify 4-1BBL/4-1BB as a downstream target of HDACi, especially of MS-275 anti-leukemia action in vitro. Thus, HDACi such as MS-275 displaying dual TNF-dependent proapoptotic and costimulatory activities might be favored for inclusion in HDACi-based anti-cancer therapeutic strategies.
Collapse
Affiliation(s)
- Bérengère Vire
- INSERM U891, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- Institut Paoli-Calmettes, Marseille, France
- Université de la Méditerranée, Marseille, France
| | - Stéphane de Walque
- Université Libre de Bruxelles, Institut de Biologie et de Médecine Moléculaires (IBMM), Laboratoire de Virologie Moléculaire, Gosselies, Belgique
| | - Audrey Restouin
- INSERM U891, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- Institut Paoli-Calmettes, Marseille, France
- Université de la Méditerranée, Marseille, France
| | - Daniel Olive
- INSERM U891, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- Institut Paoli-Calmettes, Marseille, France
- Université de la Méditerranée, Marseille, France
| | - Carine Van Lint
- Université Libre de Bruxelles, Institut de Biologie et de Médecine Moléculaires (IBMM), Laboratoire de Virologie Moléculaire, Gosselies, Belgique
| | - Yves Collette
- INSERM U891, Centre de Recherche en Cancérologie de Marseille, Marseille, France
- Institut Paoli-Calmettes, Marseille, France
- Université de la Méditerranée, Marseille, France
- * E-mail:
| |
Collapse
|
279
|
Sun P, Xia S, Lal B, Eberhart CG, Quinones-Hinojosa A, Maciaczyk J, Matsui W, Dimeco F, Piccirillo SM, Vescovi AL, Laterra J. DNER, an epigenetically modulated gene, regulates glioblastoma-derived neurosphere cell differentiation and tumor propagation. Stem Cells 2009; 27:1473-86. [PMID: 19544453 PMCID: PMC2935595 DOI: 10.1002/stem.89] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurospheres derived from glioblastoma (GBM) and other solid malignancies contain neoplastic stem-like cells that efficiently propagate tumor growth and resist cytotoxic therapeutics. The primary objective of this study was to use histone-modifying agents to elucidate mechanisms by which the phenotype and tumor-promoting capacity of GBM-derived neoplastic stem-like cells are regulated. Using established GBM-derived neurosphere lines and low passage primary GBM-derived neurospheres, we show that histone deacetylase (HDAC) inhibitors inhibit growth, induce differentiation, and induce apoptosis of neoplastic neurosphere cells. A specific gene product induced by HDAC inhibition, Delta/Notch-like epidermal growth factor-related receptor (DNER), inhibited the growth of GBM-derived neurospheres, induced their differentiation in vivo and in vitro, and inhibited their engraftment and growth as tumor xenografts. The differentiating and tumor suppressive effects of DNER, a noncanonical Notch ligand, contrast with the previously established tumor-promoting effects of canonical Notch signaling in brain cancer stem-like cells. Our findings are the first to implicate noncanonical Notch signaling in the regulation of neoplastic stem-like cells and suggest novel neoplastic stem cell targeting treatment strategies for GBM and potentially other solid malignancies.
Collapse
Affiliation(s)
- Peng Sun
- Department of Neurology, The Johns Hopkins University School of Medicine and The Kennedy Krieger Research Institute, Baltimore, Maryland, USA
| | - Shuli Xia
- Department of Neurology, The Johns Hopkins University School of Medicine and The Kennedy Krieger Research Institute, Baltimore, Maryland, USA
| | - Bachchu Lal
- Department of Neurology, The Johns Hopkins University School of Medicine and The Kennedy Krieger Research Institute, Baltimore, Maryland, USA
| | - Charles G. Eberhart
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | - Jarek Maciaczyk
- Department of General Neurosurgery and Laboratory of Molecular Neurosurgery, University of Freiburg, Freiburg, Germany
| | - William Matsui
- Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Francesco Dimeco
- Department of Neurosurgery, Instituto Nazionale Neurologico “Carlo Besta,” Milan, Italy
| | - Sara M. Piccirillo
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Milan, Italy
| | - Angelo L. Vescovi
- Department of Biotechnology and Biosciences, University of Milan Bicocca, Milan, Italy
| | - John Laterra
- Department of Neurology, The Johns Hopkins University School of Medicine and The Kennedy Krieger Research Institute, Baltimore, Maryland, USA
| |
Collapse
|
280
|
Early epigenetic changes and DNA damage do not predict clinical response in an overlapping schedule of 5-azacytidine and entinostat in patients with myeloid malignancies. Blood 2009; 114:2764-73. [PMID: 19546476 DOI: 10.1182/blood-2009-02-203547] [Citation(s) in RCA: 224] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sequential administration of DNA methyltransferase (DNMT) inhibitors and histone deacetylase (HDAC) inhibitors has demonstrated clinical efficacy in patients with hematologic malignancies. However, the mechanism behind their clinical efficacy remains controversial. In this study, the methylation dynamics of 4 TSGs (p15(INK4B), CDH-1, DAPK-1, and SOCS-1) were studied in sequential bone marrow samples from 30 patients with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) who completed a minimum of 4 cycles of therapy with 5-azacytidine and entinostat. Reversal of promoter methylation after therapy was observed in both clinical responders and nonresponders across all genes. There was no association between clinical response and either baseline methylation or methylation reversal in the bone marrow or purified CD34(+) population, nor was there an association with change in gene expression. Transient global hypomethylation was observed in samples after treatment but was not associated with clinical response. Induction of histone H3/H4 acetylation and the DNA damage-associated variant histone gamma-H2AX was observed in peripheral blood samples across all dose cohorts. In conclusion, methylation reversal of candidate TSGs during cycle 1 of therapy was not predictive of clinical response to combination "epigenetic" therapy. This trial is registered with http://www.clinicaltrials.gov under NCT00101179.
Collapse
|
281
|
Cang S, Ma Y, Liu D. New clinical developments in histone deacetylase inhibitors for epigenetic therapy of cancer. J Hematol Oncol 2009; 2:22. [PMID: 19486511 PMCID: PMC2695818 DOI: 10.1186/1756-8722-2-22] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Accepted: 06/01/2009] [Indexed: 02/08/2023] Open
Abstract
DNA methylation and histone acetylation are two well known epigenetic chromatin modifications. Epigenetic agents leading to DNA hypomethylation and histone hyperacetylation have been approved for treatment of hematological disorders. The first histone deacetylase inhibitor, vorinostat, has been licensed for cutaneous T cell lymphoma treatment. More than 11 new epigenetic agents are in various stages of clinical development for therapy of multiple cancer types. In this review we summarize novel histone deacetylase inhibitors and new regimens from clinical trials for epigenetic therapy of cancer.
Collapse
Affiliation(s)
- Shundong Cang
- Division of Hematology/Oncology, New York Medical College, Valhalla, NY 10595, USA
- Henan Province People's Hospital, Zhengzhou, PR China
| | - Yuehua Ma
- Division of Hematology/Oncology, New York Medical College, Valhalla, NY 10595, USA
- Henan Province People's Hospital, Zhengzhou, PR China
| | - Delong Liu
- Division of Hematology/Oncology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
282
|
Kang MR, Kang JS, Han SB, Kim JH, Kim DM, Lee K, Lee CW, Lee KH, Lee CH, Han G, Kang JS, Kim HM, Park SK. A novel delta-lactam-based histone deacetylase inhibitor, KBH-A42, induces cell cycle arrest and apoptosis in colon cancer cells. Biochem Pharmacol 2009; 78:486-94. [PMID: 19445901 DOI: 10.1016/j.bcp.2009.05.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 05/04/2009] [Accepted: 05/05/2009] [Indexed: 12/17/2022]
Abstract
In this study, we investigated the anti-tumor activity of KBH-A42 [N-hydroxy-3-(2-oxo-1-(3-phenylpropyl)-1,2,5,6-tetrahydropyridin-3-yl)propanamide], a novel synthetic histone deacetylase (HDAC) inhibitor. KBH-A42 inhibited a variety of HDAC isoforms in enzyme assays and suppressed growth of various cancer cell lines. Among the cell lines examined, colon cancer cells, including SW620, SW480 and HCT-15, were the cell types most sensitive to KBH-A42. KBH-A42 inhibition of cancer cell growth was comparable to or stronger than that of suberoylanilide hydroxamic acid (SAHA), a well-known HDAC inhibitor approved by the FDA to treat cutaneous T cell lymphomas. In SW620 cells, KBH-A42 increased the acetylation of histones, mediated cell cycle arrest (G1 arrest at low doses and G2 arrest at high doses), and induced apoptosis. The cell cycle arrest and apoptosis induced by KBH-A42 might be mediated through up-regulation of p21(Waf1) and activation of caspases, respectively. In addition, KBH-A42 inhibited SW620 tumor growth in a human tumor xenograft model. Taken together, our results indicate that KBH-A42 exerts an anti-tumor activity in vitro and in vivo and is a promising therapeutic candidate to treat human cancers.
Collapse
Affiliation(s)
- Moo Rim Kang
- Bioevaluation Center, Korea Research Institute of Bioscience and Biotechnology, Yangcheong, Ochang, Cheongwon, Chungbuk, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
283
|
Lee AYH, Paweletz CP, Pollock RM, Settlage RE, Cruz JC, Secrist JP, Miller TA, Stanton MG, Kral AM, Ozerova NDS, Meng F, Yates NA, Richon V, Hendrickson RC. Quantitative analysis of histone deacetylase-1 selective histone modifications by differential mass spectrometry. J Proteome Res 2009; 7:5177-86. [PMID: 19367703 DOI: 10.1021/pr800510p] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Inhibitors of class 1 and class 2 histone deacetylase (HDAC) enzymes have shown antitumor activity in human clinical trials. More recently, there has been interest in developing subtype-selective HDAC inhibitors designed to retain anticancer activity while reducing potential side effects. Efforts have been initiated to selectively target HDAC1 given its role in tumor proliferation and survival. The development of HDAC1-specific inhibitors will require the identification of HDAC1-selective pharmacodynamic markers that correlate closely with HDAC1-inhibition in vitro and in vivo. Existing histone markers of HDAC target engagement were developed using pan-HDAC inhibitors and do not necessarily represent robust readouts for isoform-specific inhibitors. Therefore, we have initiated a proteomic approach to identify readouts for HDAC1 inhibition. This approach involves the use of differential mass spectrometry (dMS) to identify post-translational changes in histones by profiling histone-enriched cellular fractions treated with various HDAC inhibitors. In this study, we profiled histones isolated from the HCT116 human colon cancer cell line that have been treated with compounds from multiple chemical classes that are specific for HDAC1; HDAC1 and 3; and HDAC1, 3, and 6 enzymes. In two independent experiments, we identified 24 features that correlated with HDAC1-inhibition. Among the peptides modulated by HDAC1-selective inhibitors were Ac-H2B-K5 from histone H2B, and Ac-H3-K18 from histone H3. Commercially available antibodies to specific histone acetyl-lysine residues were used to confirm that these peptides also provide pharmacodynamic readouts for HDAC1-selective inhibitors in vivo and in vitro. These results show the utility of dMS in guiding the identification of specific readouts to aid in the development of HDAC-selective inhibitors.
Collapse
Affiliation(s)
- Anita Y H Lee
- Department of Proteomics, Merck Research Laboratories, Merck & Co. Inc., Rahway, New Jersey 07065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
284
|
Epigenetic influences on sensory regeneration: histone deacetylases regulate supporting cell proliferation in the avian utricle. J Assoc Res Otolaryngol 2009; 10:341-53. [PMID: 19340485 DOI: 10.1007/s10162-009-0166-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 02/16/2009] [Indexed: 10/20/2022] Open
Abstract
The sensory hair cells of the cochlea and vestibular organs are essential for normal hearing and balance function. The mammalian ear possesses a very limited ability to regenerate hair cells and their loss can lead to permanent sensory impairment. In contrast, hair cells in the avian ear are quickly regenerated after acoustic trauma or ototoxic injury. The very different regenerative abilities of the avian vs. mammalian ear can be attributed to differences in injury-evoked expression of genes that either promote or inhibit the production of new hair cells. Gene expression is regulated both by the binding of cis-regulatory molecules to promoter regions as well as through structural modifications of chromatin (e.g., methylation and acetylation). This study examined effects of histone deacetylases (HDACs), whose main function is to modify histone acetylation, on the regulation of regenerative proliferation in the chick utricle. Cultures of regenerating utricles and dissociated cells from the utricular sensory epithelia were treated with the HDAC inhibitors valproic acid, trichostatin A, sodium butyrate, and MS-275. All of these molecules prevent the enzymatic removal of acetyl groups from histones, thus maintaining nuclear chromatin in a "relaxed" (open) configuration. Treatment with all inhibitors resulted in comparable decreases in supporting cell proliferation. We also observed that treatment with the HDAC1-, 2-, and 3-specific inhibitor MS-275 was sufficient to reduce proliferation and that two class I HDACs--HDAC1 and HDAC2--were expressed in the sensory epithelium of the utricle. These results suggest that inhibition of specific type I HDACs is sufficient to prevent cell cycle entry in supporting cells. Notably, treatment with HDAC inhibitors did not affect the differentiation of replacement hair cells. We conclude that histone deacetylation is a positive regulator of regenerative proliferation but is not critical for avian hair cell differentiation.
Collapse
|
285
|
Abstract
Tumorigenesis is traditionally thought to be caused by the imbalance between oncogenes and tumor-suppressor genes. Epigenetics is a recently described phenomenon that uses an alternative mechanism to explain the transcriptional inactivation of tumor-suppressor genes predominantly by hypermethylation of the promoter regions. Hypermethylation of these regions has been described extensively in many neoplasms, including cutaneous melanoma. Histone modification, primarily by acetylation and deacetylation, is a current potential target for melanoma therapy, but more research is required to understand the mechanisms involved and the therapeutic effectiveness of regimens involving these agents. These mechanisms not only are important for understanding the origin and progression of neoplasms but also have important potential therapeutic implications. Understanding the epigenetic mechanisms involved in melanoma can provide valuable information with significant implications in diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Willmar D Patino
- Department of Pathology, University of Texas Southwestern Medical Center in Dallas, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | | |
Collapse
|
286
|
McCulloch MW, Coombs GS, Banerjee N, Bugni TS, Cannon KM, Harper MK, Veltri CA, Virshup DM, Ireland CM. Psammaplin A as a general activator of cell-based signaling assays via HDAC inhibition and studies on some bromotyrosine derivatives. Bioorg Med Chem 2009; 17:2189-98. [PMID: 19022675 PMCID: PMC2670876 DOI: 10.1016/j.bmc.2008.10.077] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Accepted: 10/31/2008] [Indexed: 01/21/2023]
Abstract
The Wnt signaling pathway regulates cell growth and development in metazoans, and is therefore of interest for drug discovery. By screening a library of 5808 pre-fractionated marine extracts in a cell-based Wnt signaling assay, several signaling activators and inhibitors were observed. LCMS-based fractionation rapidly identified an active compound from Pseudoceratina purpurea as psammaplin A, a known HDAC inhibitor. Other HDAC inhibitors similarly activated signaling in this assay, indicating HDAC inhibitors will be identified through many cell-based reporter assays. In a large scale analysis of P. purpurea, three previously undescribed bromotyrosine based natural products were identified; the structure of one of these was confirmed by synthesis. Additionally, three other derivatives of psammaplin A were prepared: a mixed disulfide and two sulfinate esters. Finally, evidence to support a structural reassignment of psammaplin I from a sulfone to the isomeric sulfinate ester is presented.
Collapse
Affiliation(s)
| | | | - Nikhil Banerjee
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Tim S. Bugni
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, USA
| | - Kendell M. Cannon
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Mary Kay Harper
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, USA
| | - Charles A. Veltri
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, USA
| | | | - Chris M. Ireland
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
287
|
Wang H, Yu N, Song H, Chen D, Zou Y, Deng W, Lye PL, Chang J, Ng M, Blanchard S, Sun ET, Sangthongpitag K, Wang X, Goh KC, Wu X, Khng HH, Fang L, Goh SK, Ong WC, Bonday Z, Stünkel W, Poulsen A, Entzeroth M. N-Hydroxy-1,2-disubstituted-1H-benzimidazol-5-yl acrylamides as novel histone deacetylase inhibitors: Design, synthesis, SAR studies, and in vivo antitumor activity. Bioorg Med Chem Lett 2009; 19:1403-8. [DOI: 10.1016/j.bmcl.2009.01.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 01/09/2009] [Accepted: 01/13/2009] [Indexed: 11/26/2022]
|
288
|
Raeppel S, Zhou N, Gaudette F, Leit S, Paquin I, Larouche G, Moradei O, Fréchette S, Isakovic L, Delorme D, Fournel M, Kalita A, Lu A, Trachy-Bourget MC, Yan PT, Liu J, Rahil J, Wang J, Besterman JM, Murakami K, Li Z, Vaisburg A. SAR and biological evaluation of analogues of a small molecule histone deacetylase inhibitor N-(2-aminophenyl)-4-((4-(pyridin-3-yl)pyrimidin-2-ylamino)methyl)benzamide (MGCD0103). Bioorg Med Chem Lett 2009; 19:644-9. [DOI: 10.1016/j.bmcl.2008.12.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 12/11/2008] [Accepted: 12/12/2008] [Indexed: 11/28/2022]
|
289
|
Parallel medicinal chemistry approaches to selective HDAC1/HDAC2 inhibitor (SHI-1:2) optimization. Bioorg Med Chem Lett 2009; 19:1168-72. [DOI: 10.1016/j.bmcl.2008.12.083] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Revised: 12/17/2008] [Accepted: 12/19/2008] [Indexed: 11/21/2022]
|
290
|
Caspase-8 dependent histone acetylation by a novel proteasome inhibitor, NPI-0052: a mechanism for synergy in leukemia cells. Blood 2009; 113:4289-99. [PMID: 19182209 DOI: 10.1182/blood-2008-08-174797] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Combination studies of histone deacetylase inhibitors (HDACi) and proteasome inhibitors are providing preclinical framework to build better strategies against hematologic malignancies. Our previous work found that a novel proteasome inhibitor, NPI-0052, and HDACi synergistically induce apoptosis in leukemia cells in a caspase-8- and oxidant-dependent manner. Here we extend those observations to primary leukemia cells and identify novel mechanisms of synergy. Because the proximal targets of NPI-0052 and HDACi are inhibition of proteasome activity and histone acetylation, we initially examined those biochemical events. Increased acetylation of histone-H3 was detected in Jurkat and CLL primary cells treated with NPI-0052, alone or in combination with various HDACi (MS/SNDX-275 or vorinostat). Hyperacetylation by NPI-0052 occurred to a lesser extent in caspase-8-deficient cells and in cells treated with an antioxidant. These results indicate that NPI-0052 is eliciting caspase-8 and oxidative stress-dependent epigenetic alterations. In addition, real-time PCR revealed that MS/SNDX-275 repressed expression of the proteasomal beta5, beta2, and beta1 subunits, consequently inhibiting respective enzymatic activities. Overall, our results suggest that crosstalk by NPI-0052 and HDACi are contributing, along with caspase-8 activation and oxidative stress, to their synergistic cytotoxic effects in leukemia cells, reinforcing the potential clinical utility of combining these 2 agents.
Collapse
|
291
|
Danish M, Gardner ER, Chen X, Figg WD. Determination of a benzamide histone deacetylase inhibitor, MS-275, in human plasma by liquid chromatography with mass-spectrometric detection. J Chromatogr B Analyt Technol Biomed Life Sci 2009; 877:355-9. [PMID: 19117815 PMCID: PMC2661872 DOI: 10.1016/j.jchromb.2008.12.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 11/24/2008] [Accepted: 12/08/2008] [Indexed: 11/24/2022]
Abstract
An analytical method was developed and validated for the quantitative determination of the histone deacetylase inhibitor MS-275 in human plasma. Calibration curves were linear in the concentration range of 1-250 ng/mL. Sample pretreatment involved a liquid-liquid extraction of 0.1 mL aliquots of plasma with methyl tert-butyl ether. MS-275 and the internal standard, benzanilide, were separated on a Zorbax SB-Phenyl column (4.6 mm x 75 mm I.D., 3.5 microm), using a mobile phase composed of methanol and 10 mM ammonium formate (pH 2.9). The column eluent was monitored by mass spectrometry with electrospray ionization. Accuracy and precision of three concentrations of quality control samples ranged from 96.56 to 107.02% and 0.97 to 4.29%, respectively. This method represents an improvement over the previously published analytical assay for this agent, increasing the accuracy and precision (through addition of a suitable internal standard) and expanding the analytical range. The developed method was applied to study the pharmacokinetics of MS-275 in 724 clinical samples.
Collapse
Affiliation(s)
- Matthew Danish
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Erin R. Gardner
- Clinical Pharmacology Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD 21702, USA
| | - Xiaohong Chen
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
292
|
Marquard L, Gjerdrum LM, Christensen IJ, Jensen PB, Sehested M, Ralfkiaer E. Prognostic significance of the therapeutic targets histone deacetylase 1, 2, 6 and acetylated histone H4 in cutaneous T-cell lymphoma. Histopathology 2009. [PMID: 18671804 PMCID: PMC2675007 DOI: 10.1111/j.1365-2559.2008.03109.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AIMS Aberrant histone acetylation has been associated with malignancy and histone deacetylase (HDAC) inhibitors are currently being investigated in numerous clinical trials. So far, the malignancy most sensitive to HDAC inhibitors has been cutaneous T-cell lymphoma (CTCL). The reason for this sensitivity is unclear and studies on HDAC expression and histone acetylation in CTCL are lacking. The aim of this study was to address this issue. METHODS AND RESULTS The immunohistochemical expression of HDAC1, HDAC2, HDAC6, and acetylated H4 was examined in 73 CTCLs and the results related to histological subtypes and overall survival. HDAC1 was most abundantly expressed (P < 0.0001), followed by HDAC2; HDAC6 and H4 acetylation were equally expressed. HDAC2 (P = 0.001) and H4 acetylation (P = 0.03) were significantly more common in aggressive than indolent CTCL subtypes. In contrast, no differences were observed for HDAC1 and HDAC6. In a Cox analysis, elevated HDAC6 was the only parameter showing significant influence on survival (P = 0.04). CONCLUSIONS High expression of HDAC2 and acetylated H4 is more common in aggressive than indolent CTCL. HDAC6 expression is associated with a favorable outcome independent of the subtype.
Collapse
Affiliation(s)
- L Marquard
- Department of Pathology, Experimental Pathology Unit, Copenhagen Biocentre, Copenhagen University Hospital, Copenhagen.
| | | | | | | | | | | |
Collapse
|
293
|
Schmidt K, Seeger K, Scheibenbogen C, Bender R, Abdulla M, Süssmilch S, Salama A, Moldenhauer A. Histone deacetylase inhibition improves differentiation of dendritic cells from leukemic blasts of patients with TEL/AML1-positive acute lymphoblastic leukemia. J Leukoc Biol 2009; 85:563-73. [DOI: 10.1189/jlb.0808469] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
294
|
Multicenter phase II trial of the histone deacetylase inhibitor pyridylmethyl-N-{4-[(2-aminophenyl)-carbamoyl]-benzyl}-carbamate in pretreated metastatic melanoma. Melanoma Res 2009; 18:274-8. [PMID: 18626312 DOI: 10.1097/cmr.0b013e328307c248] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Systemic treatment of metastatic melanoma is of low efficacy, and new therapeutic strategies are needed. Histone deacetylase inhibitors are supposed to restore the expression of tumor suppressor genes and induce tumor cell differentiation, growth arrest, and apoptosis. This study was aimed to evaluate the efficacy, safety, and pharmacokinetics of the histone deacetylase inhibitor pyridylmethyl-N-{4-[(2-aminophenyl)-carbamoyl]-benzyl}-carbamate (MS-275) in patients with pretreated metastatic melanoma. Patients with unresectable AJCC stage IV melanoma refractory to at least one earlier systemic therapy were randomized to receive MS-275 3 mg biweekly (days 1+15, arm A) or 7 mg weekly (days 1+8+15, arm B), in 4-week cycles. The primary study endpoint was objective tumor response, secondary endpoints were safety and time-to-progression. On the basis of Simon's two-stage design, the study initially allowed an entry of 14 patients per arm; if there was at least one responder, additional 33 patients were to be enrolled. Among 28 patients enrolled, no objective response was detected. Four (29%) patients in arm A and three (21%) patients in arm B showed disease stabilizations. Median time-to-progression was comparable in both arms with 55.5 versus 51.5 days, respectively; median overall survival was 8.84 months. Toxicity was mild to moderate with nausea (39%) and hypophosphatemia (29%) as the most frequently reported events. No treatment-related serious adverse events occurred. Single-agent treatment with MS-275 was well-tolerated and showed long-term tumor stabilizations, but no objective responses in pretreated metastatic melanoma. Further evaluation of MS-275 in combination schedules is warranted.
Collapse
|
295
|
Jain N, Rossi A, Garcia-Manero G. Epigenetic therapy of leukemia: An update. Int J Biochem Cell Biol 2009; 41:72-80. [PMID: 18948224 PMCID: PMC3833715 DOI: 10.1016/j.biocel.2008.10.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 10/13/2008] [Accepted: 10/13/2008] [Indexed: 02/03/2023]
Abstract
Carcinogenesis is classically thought to result from genetic alterations in DNA sequence such as deletions, mutations, or chromosomal translocations. These in turn may lead to the activation of oncogenes, inactivation of tumor suppressor genes or formation of chimeric oncoproteins. Epigenetics, in contrast, refers to a number of biochemical modifications of chromatin, either to DNA directly or to its associated protein complexes that affect gene expression without altering the primary sequence of DNA [Robertson KD, Wolffe AP. DNA methylation in health and disease. Nat Rev Genet 2000;1:11-9; Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128:683-92]. A fundamental difference between genetic and epigenetic alterations is the irreversible nature of genetic lesions whereas epigenetic ones are potentially reversible, allowing for therapeutic intervention. In the last decade, it has become apparent that epigenetic changes play an important role in cancer, particularly in leukemia. Significant advances have been made in the elucidation of these processes as well as in translating this knowledge to the clinic, as in the development of new prognostic biomarkers or targeted therapies. In this review, we will focus on recent advances in epigenetic therapy in leukemia.
Collapse
Affiliation(s)
- Nitin Jain
- Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Adriana Rossi
- Department of Medicine, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030 USA
| |
Collapse
|
296
|
Kwon HK, Ahn SH, Park SH, Park JH, Park JW, Kim HM, Park SK, Lee K, Lee CW, Choi E, Han G, Han JW. A Novel .GAMMA.-Lactam-Based Histone Deacetylase Inhibitor Potently Inhibits the Growth of Human Breast and Renal Cancer Cells. Biol Pharm Bull 2009; 32:1723-7. [DOI: 10.1248/bpb.32.1723] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Seong Hoon Ahn
- Division of Molecular & Life Science, Hanyang University
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
297
|
Induction of Foxp3+ regulatory T cells with histone deacetylase inhibitors. Cell Immunol 2009; 257:97-104. [DOI: 10.1016/j.cellimm.2009.03.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 03/07/2009] [Accepted: 03/10/2009] [Indexed: 12/23/2022]
|
298
|
Kuhn AN, van Santen MA, Schwienhorst A, Urlaub H, Lührmann R. Stalling of spliceosome assembly at distinct stages by small-molecule inhibitors of protein acetylation and deacetylation. RNA (NEW YORK, N.Y.) 2009; 15:153-75. [PMID: 19029308 PMCID: PMC2612777 DOI: 10.1261/rna.1332609] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The removal of intervening sequences from a primary RNA transcript is catalyzed by the spliceosome, a large complex consisting of five small nuclear (sn) RNAs and more than 150 proteins. At the start of the splicing cycle, the spliceosome assembles anew onto each pre-mRNA intron in an ordered process. Here, we show that several small-molecule inhibitors of protein acetylation/deacetylation block the splicing cycle: by testing a small number of bioactive compounds, we found that three small-molecule inhibitors of histone acetyltransferases (HATs), as well as three small-molecule inhibitors of histone deacetylases (HDACs), block pre-mRNA splicing in vitro. By purifying and characterizing the stalled spliceosomes, we found that the splicing cycle is blocked at distinct stages by different inhibitors: two inhibitors allow only the formation of A-like spliceosomes (as determined by the size of the stalled complexes and their snRNA composition), while the other compounds inhibit activation for catalysis after incorporation of all U snRNPs into the spliceosome. Mass-spectrometric analysis of affinity-purified stalled spliceosomes indicated that the intermediates differ in protein composition both from each other and from previously characterized native A and B splicing complexes. This suggests that the stalled complexes represent hitherto unobserved intermediates of spliceosome assembly.
Collapse
Affiliation(s)
- Andreas N Kuhn
- Department of Cellular Biochemistry, Max Planck Institute for Biophysical Chemistry, D-37077 Göttingen, Germany
| | | | | | | | | |
Collapse
|
299
|
Chittur SV, Sangster-Guity N, McCormick PJ. Histone deacetylase inhibitors: a new mode for inhibition of cholesterol metabolism. BMC Genomics 2008; 9:507. [PMID: 18959802 PMCID: PMC2613157 DOI: 10.1186/1471-2164-9-507] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Accepted: 10/29/2008] [Indexed: 11/12/2022] Open
Abstract
Background Eukaryotic gene expression is a complex process involving multiple cis and trans activating molecules to either facilitate or inhibit transcription. In recent years, many studies have focused on the role of acetylation of histone proteins in modulating transcription, whereas deacetylation of these same proteins is associated with inactivation or repression of gene expression. This study explores gene expression in HepG2 and F9 cell lines treated with Trichostatin A (TSA), a potent histone deacetylase inhibitor. Results These experiments show that TSA treatment results in clear repression of genes involved in the cholesterol biosynthetic pathway as well as other associated pathways including fatty acid biosynthesis and glycolysis. TSA down regulates 9 of 15 genes in this pathway in the F9 embryonal carcinoma model and 11 of 15 pathway genes in the HepG2 cell line. A time course study on the effect of TSA on gene expression of various enzymes and transcription factors involved in these pathways suggests that down regulation of Srebf2 may be the triggering factor for down regulation of the cholesterol biosynthesis pathway. Conclusion Our results provide new insights in the effects of histone deacetylases on genes involved in primary metabolism. This observation suggests that TSA, and other related histone deacetylase inhibitors, may be useful as potential therapeutic entities for the control of cholesterol levels in humans.
Collapse
Affiliation(s)
- Sridar V Chittur
- Center for Functional Genomics, University at Albany, State University of New York, Cancer Research Center, One Discovery Drive, Rm 310, Rensselaer, NY 12144, USA.
| | | | | |
Collapse
|
300
|
Chou CJ, Herman D, Gottesfeld JM. Pimelic diphenylamide 106 is a slow, tight-binding inhibitor of class I histone deacetylases. J Biol Chem 2008; 283:35402-9. [PMID: 18953021 DOI: 10.1074/jbc.m807045200] [Citation(s) in RCA: 177] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Histone deacetylase (HDAC) inhibitors, including various benzamides and hydroxamates, are currently in clinical development for a broad range of human diseases, including cancer and neurodegenerative diseases. We recently reported the identification of a family of benzamide-type HDAC inhibitors that are relatively non-toxic compared with the hydroxamates. Members of this class of compounds have shown efficacy in cell-based and mouse models for the neurodegenerative diseases Friedreich ataxia and Huntington disease. Considerable differences in IC(50) values for the various HDAC enzymes have been reported for many of the HDAC inhibitors, leading to confusion as to the HDAC isotype specificities of these compounds. Here we show that a benzamide HDAC inhibitor, a pimelic diphenylamide (106), is a class I HDAC inhibitor, demonstrating no activity against class II HDACs. 106 is a slow, tight-binding inhibitor of HDACs 1, 2, and 3, although inhibition for these enzymes occurs through different mechanisms. Inhibitor 106 also has preference toward HDAC3 with K(i) of approximately 14 nm, 15 times lower than the K(i) for HDAC1. In comparison, the hydroxamate suberoylanilide hydroxamic acid does not discriminate between these enzymes and exhibits a fast-on/fast-off inhibitory mechanism. These observations may explain a paradox involving the relative activities of pimelic diphenylamides versus hydroxamates as gene activators.
Collapse
Affiliation(s)
- C James Chou
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | |
Collapse
|