Accepted Manuscript Title: Histone modifications: a review about the presence of this epigenetic phenomenon in carcinogenesis Authors: Emanuely Silva Chrun, Filipe Modolo, Filipe Ivan Daniel PII: DOI: Reference:
S0344-0338(17)30318-7 http://dx.doi.org/doi:10.1016/j.prp.2017.06.013 PRP 51832
To appear in: Received date: Revised date: Accepted date:
3-4-2017 23-5-2017 24-6-2017
Please cite this article as: Emanuely Silva Chrun, Filipe Modolo, Filipe Ivan Daniel, Histone modifications: a review about the presence of this epigenetic phenomenon in carcinogenesis, Pathology - Research and Practicehttp://dx.doi.org/10.1016/j.prp.2017.06.013 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Histone modifications: a review about the presence of this epigenetic phenomenon in carcinogenesis Emanuely Silva Chrun a, Filipe Modolo b, Filipe Ivan Daniel b,* a
DDS, MSc, PhD student of Post graduate program in Dentistry-Oral Diagnostic, Department of Pathology, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil. E-mail:
[email protected] b DDS, PhD, Professor Department of Pathology, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil. E-mail:
[email protected],
[email protected]. * Correspondence may also be addressed to Filipe Ivan Daniel. Tel: +55 48 3721 3483; Fax: +55 48 3721 9473; Email:
[email protected]
Abstract Among the epigenetic changes, histone acetylation has been recognized as a fundamental process that strongly affects gene expression regulation. Disrupt of this phenomenon has been linked to carcinogenesis. In this review, we analysed studies reporting the process of histone modification, the enzymes associated and affected genes concerning human malignancies and histone enzyme inhibitor drugs used in cancer treatment. Variable degrees of expression of HDACs (histone deacetylases) and HATs (histone acetyltransferases) are found in many human malignant tissues and the histones acetylation seems to influence different processes including the progression of cell cycle, the dynamics of chromosomes, DNA recombination, DNA repair and apoptosis. Thus, the control of aberrant activity and/or expression of these proteins have been favorable in treatment of diseases as cancer. HDACi have shown efficacy in clinical trials in solid and hematological malignancies. Therefore, the development and use of HDACs inhibitors are increasing, leading to continue studying these enzyme expressions and behavior, aiming to determine tumors that will respond better to this type of treatment. Keywords: Histone Deacetylases; ; ; , Histone Acetyltransferases, Genes, Review. Introduction Epigenetic is an interesting and relatively recent field in biology, referring to the regulation of gene expression, without alterations in DNA sequence and with a heritable pattern during cell division. Progress has been made in knowledge of epigenetic modifications in normal and diseased tissues ` [29, 111]. It is known that these modifications are reversible and play a key role in develop ment of some disorders[150]. Researches in epigenetics have provided new insights into some kinds of diseases, especially human malignancies, neurodevelopmental disorders (Alzheimer's, Parkinson's and Huntington's diseases, multiple sclerosis and epilepsy)[139] and autoimmune sickness (rheumatoid arthritis[59], type 1 diabetes[93] and lupus erythematosus[55]). Besides the already known genetic alterations, the epigenetic deregulation is of great importance in the development of malignant tumors[23] becoming an obvious focus of research in this field[94, 111]. The main types of epigenetic information are: a) DNA methylation where the addition of a methyl group to 5-carbon of cytosine in CpG islands[17] promotes gene silencing and also involves histone deacetylation through interaction with co-repressor complexes[102]; b) miRNAs[58, 75], small noncoding RNAs involved in the regulation of fundamental cellular processes and crucial to transcription and translation[36], which deregulation have been related to many human cancers pathogenesis[64]; and, c) histone modifications, characterized mainly by alterations in the acetylation status, also providing access or not of transcription factors to sequences of nucleotides.[22] This literature review aimed to analyze studies reporting the process of histone modification, the enzymes related and affected genes concerning human malignancies as well histone deacetylase inhibitors (HDACi) drugs already used for cancer treatment. Histones, histone modifications and related enzymes Histones are proteins that, together with DNA, comprises the nucleosome formed by the core histone octamer (two heterodimers of H2A and H2B along with two heterodimers of H3 and H4), which
is wrapped by two turns of 147-base-paired DNA strand[106], while the H1 protein establishes and maintains the higher-order chromatin structures[53]. Each histone comprising the octamer within the nucleosome is rich in lysine tails extending out of it and, therefore, the accessibility to the DNA in the nucleosome is, in part, controlled by modifications of these tails[130]. Histones may be submitted to several post-translational modifications as ribosylation, ubiquitynation and sumoylation of lysines, phosphorylation of serines and threonines, acetylation or methylation of lysines and arginines. Lysine residues in histone tails have a positive charge that interacts with the negatively charged phosphate backbone of DNA. Covalent modifications lead to alterations in chromatin structure and, thus, affect accessibility of transcription factors to ‘read’ and/or copy the nucleotide base sequences; therefore, these structures are dynamic and have capacity of folding (heterochromatin) and unfolding (euchromatin) regulation[106]. Histone deacetylases (HDACs) and histone acetyltransferases (HATs) are enzymes that influence DNA transcription through the balance between histone acetylation and deacetylation in normal cell function. Histone acetylation reduces chromatin condensation (Fig. 1): HATs catalyse the transference of an acetyl group (negatively charged) from acetyl-CoA to the amino-terminal tail of lysine, neutralizing the histone positive charge[12]. It generates slackness between DNA-histone and also between nucleosomes, allowing access of transcription factors and, therefore, DNA transcription. On the other hand, histone deacetylation returns to original condensation: HDACs remove acetyl group recovering the positive charge, permitting interactions between negatively charged DNA and histone protein, resulting in condensation of chromatin structure (Fig. 2), which is associated with gene repression[91, 94]. To date, 18 human HDACs have been identified and are classified into four classes[34, 78, 123]. Class I comprises HDAC1, HDAC2, HDAC3, and HDAC8, mainly located in the cell nucleus[38, 78]; Class II HDACs are located both in the nucleus and cytoplasm (HDAC4, 5, 7, and 9) or only in cytoplasm (HDAC6 and 10); Class IV includes HDAC11, which appears both in the nucleus and cytoplasm; while Class III is made up by Sirtuins which, different from the others histone deacetylases, are Zn-independent[78]. As HDAC1 and HDAC2 are inactive when produced by recombinant techniques, cofactors are necessary for its activity to occur. In vivo, its activity is triggered only within a complex of proteins related to HDACs[24]. Among them, the corepressor protein Sin3 is postulated to mediate protein-protein interactions in several regulatory systems of gene expression. As Sin3 does not display DNA binding domain, promoter targeting is achieved through interactions with other sequence-specific DNA-binding proteins, and therefore require HDACs as prosecutor of this link to achieve its repression function. MadMax complex is strongly induced during cell differentiation and acts suppressing cell proliferation, through the complex comprised with Sin3/HDAC[44] (Fig. 3a). Ume6, Ncor and YY1[76] (all transcription repressors) both request Sin3 that, in turn, needs HDACs to bind DNA. MeCP2 is one of the four main proteins with a methyl-CpG binding domain able to link to methylated DNA, resulting in transcriptional silencing probably due to Sin3/HDAC complex recruitment to achieve gene repression[9, 23] (Fig. 3b). Thus, HDACs indirectly or directly are requested by these other factors, enhancing its role and importance in gene silencing, showing to be fundamental for regulatory mechanisms governing cell proliferation and differentiation[39]. HATs have been organized in two general classes (A-type and B-type) based in their probable origin and function in cells. A-type, with nuclear localization, probably participates catalyzing the acetylation and events related to gene transcription. B-type HATs are likely responsible for catalyzing acetylation events related to the transport of newly synthesized histones from the cytoplasm to the nucleus and its deposition on the replicated DNA. HAT domains motif A contains binding sites to acetylCoA and are present in structure of all proteins classified as HAT activity[117]. In addition, based on its primary-structure, HATs are organized into families. The four HAT families most studied are: the GNAT family (Gcn5, PCAF, Hat1, Elp3, and Hpa2); the p300/CBP family (p300, and CBP); the MYST family (Esa1, MOF, Sas2, Sas3, MORF, Tip60, and Hbo1), and the Rtt109 [25, 117].
Many of HATs identified to date do not work isolated in vivo, and likewise HDACs, HATs often act within a coactivator complex. An in vitro study by immunoprecipitation test using antibody to recognize hyperacetylated histones noted that these histones are globally located in regions of DNase sensitivity which is a region transcriptionally active, reinforcing the role of HATs in transcription[117]. Each HAT exhibits site and histone specificity, and for each distinct physiological function specific histone acetylation at specific locations are needed. The specificity becomes wide within complexes[106]. Some of coactivators complexes most cited in studies are: ADA (Gnc5, Ada2, and Ada3), SAGA (SPT, ADA2B, ADA3, SGF29/STAF36, and GCN5/PCAF)[37, 95], STAGA (SPT3-TAF9GCN5[152]), and ATAC (Ada Two-A containing-GCN5, ADA2A, ADA3, and SGF29[143],37). GCN5 and PCAF are two paralogous yeast Gcn5-like acetyltransferases that influence diverse biological processes by acetylating histones and non-histone proteins, chromatin regulation, and gene-specific transcription as part of these multiprotein complexes[71, 143]. Within ADA and SAGA complexes, acetylation activity of Gcn5 is increased and modifies chromatin structure and regulation of the basal transcription machinery[115]. Direct interactions between histone modifying enzymes and DNA sequence-specific transcriptional regulators are crucial for achieving targeting histone modification[106]. Aberrant histone deacetylation is responsible for transcriptional repression of genes involved in negative regulation of cell differentiation and proliferation, migration and metastasis in human malignances[35]. The global histone modification levels are predictive of cancer recurrence and this indicates that histone acetylation/deacetylation is a versatile regulatory event which plays a key role in various cellular processes[124]. HDACs and HATs in cancer Although acetylation/deacetylation events are specific and well controlled processes in the normal cell, histone hipoacetylation plays an important role in cancer initiation by changing gene expression and cell phenotype[67]. Three situations are possible for its occurrence: HDACs overexpression and/or exceeded activity; decreased amount of HATs and/or its activity; and both conditions concomitantly[134]. The sequence of histone modifying enzyme action also affects the overall protein machinery: one modification leads or inhibits the other. The enzyme-substrate specificity (the specific residue of certain histone tail) is as important as the recruitment of the enzyme at the exact moment. These phenomena are associated with distinct chromatin status and, as previously discussed, alterations in chromatin status lead to changes in transcriptional process[106]. HDAC expression has been investigated in different human malignancies by several authors. In this review, we raised manuscripts about expression of HDACs (Class I, II, III and IV) in cancer tissues such as oral, breast, gastric, lung, prostate, colorectal, ovary, endometrial, pancreatic, thyroid, esophageal, leukemia, and lymphoma (Table 1). HDAC1 and HDAC2 are the deacetylases proteins most studied and well understood in human cancer and cell lines, with predominantly nuclear staining when conducting immunohistochemistry[1, 15, 42, 45, 51, 72, 73, 81, 98, 100, 121, 134, 135, 146]. Although increased expression was found in most investigations, some tumors (breast cancer[73, 98, 125] and leukemia[96]) showed inconstant or varied expression. Breast cancer and leukemia are the most investigated tissue by most authors. Breast cancer showed wide variation of HDAC1, HDAC2, and HDAC3 expression while these proteins are overexpressed in the other investigated malignancies. Leukemia[40, 96, 149] reports enhanced expression for most deacetylases types. Other authors found heterogeneous expressions within several hematologic malignancy subtypes. Abnormal expression of HDACs and HATs may cause epigenetic alterations associated with malignant cell behavior and has been associated with tumor development and progression[34, 145, 146]. Histone acetylation and deacetylation affect several gene expression involved in tumorigenesis. The HDACs may repress transcription of differentiation factors, allowing cell proliferation without differentiation, cyclin-dependent kinases inhibitors, and pro-apoptotic factors, not admitting the programmed cell death. This repression promotes the unregulated cell growth observed in the initiation and progression of malignant tumors[35]. All these events can be supported by evidence of studies that utilized HDAC inhibitors (Table 2).
The role of histone modification proteins in cancer is based on the following explanation: when there is overexpression of HDACs, probably activities of tumor suppressor genes such as p21 (cell cycle progression inhibitor) are silenced leading to tumor initiation and/or progression, even as other experiments show that deacetylation of p53 by HDACs is likely to be part of the mechanisms that control the physiological activity of p53 and depends on the p53 region acetylated by p300/CBP (HAT) [57]. It has also been suggested that loss of HDAC expression, together with the lack of control in oncogenes expression, might help tumorigenesis depending on the corepressor complex to which it binds, for example, Rb expression requires HDAC1 to repress transcription and plays a role on tumor suppressor[11, 84], the same process occurs with AML1-ETO (frequently altered in leukemias)[3]. Beyond HDAC expression changes in several types of human malignances, translocations, mutations and deletions in HAT and HAT-related genes may also contribute to global imbalance of histone acetylation[111]. With the acetylation it is suggested that the acetyl-lysine in histone tails provide recognition of the binding sites by factors involved in gene transcription[132]. The role of various types of HAT within the malignant tumors can be subject of extensive discussion, as there are evidences of its action collaborating with expression of classical tumor suppressor gene and, in turn activating oncogenes, depending on the studied target gene (Table 3). Thus, it is possible to suggest that genetic changes may lead to epigenetic changes. And this may also be the reason for the overexpression of HDACs, since some translocations result in overexpression of proto-oncogenes by removing its normal regulatory elements and keeping them under the control of a highly active promoter[74]. Thereby, histone modifying enzymes and complexes are involved in such diverse processes as transcription activation or gene silencing of key genes such as DNA repair and cell cycle regulatory genes. HDAC inhibitors (HDACi) HDACi are a group of drugs with different structures and biological activity and specificity shown to be potent antiproliferative agents, with relatively little effect on normal tissues and clinically welltolerated[63, 77]. These drugs overall exert their anticancer activity by favoring the acetylation and reversing the excess histone deacetylation leading to re-expression of silenced genes and contributing to reverse the malignant phenotype[97]. The effects of HDACi in gene transcription are complex. Among their properties are cell cycle arrest, generation of reactive oxygen species, apoptosis induction, and antiangiogenic effects. These cascades of changes in gene expression occur due to acetylation of histone and non-histone[97] nucleossomal proteins[114]. The effects in acetylation of the non-histone proteins can affect many vital regulatory process including gene transcriptional activation, resuming the production of proteins (i.e. p53, p21 and tubulin)[77]. Another important effect is that HDACi also act in cellular cycle checkpoints, allowing greater specificity for tumor cells, considering that checkpoints are often defective in cancer cells[144]. Essentially formed by zinc-binding domain, a terminal group, and a chain linker between them, synthetic HDACi are classified according to their distinct chemical structures in mainly four classes[28]:
Hidroxamic acid-based (related to trichostatin A [TSA], from the Streptomyces hygroscopicus): vorinostat (SAHA), belinostat, abexinostat, pracinostat, resminostat, givinostat, panobinostat, and CUDC-101; Benzamide derivatives: mocetinostat, entinostat and tacedinaline Short chain fatty acid-based (related to sodium butirate): valproic acid and phenylbutyrate Cyclic peptide: romidepsin Several inhibitors have been developed and are in clinical trials to date. HDACi have been tested in solid and hematological tumors showing good results. Nevertheless, a study by Haberland et al. (2009)[41] showed that systemic administration of HDACi, which can inhibit most or all HDACs ubiquitously expressed, is well tolerated in vivo and acts upon numerous disease-associated gene expression programs in an apparently specific manner. Thereby unexpectedly, normal cells are often less sensitive to HDACi than are tumor cells[41], which is very positive in terms of antineoplastic therapy. The use of inhibitors has been clinically validated in cancer patients by FDA (Food and Drug
Administration, USA) approval. Moreover, clinical trials of several HDACi for use as anticancer drugs alone or in combination with other therapeutic agents are in progress (Table 4). HATs and HDACs are recognized as fundamental in the process that affect strongly the regulation of gene expression. These structural and functional studies have supported efforts to elucidate the molecular characteristics, mechanisms and regulation of these enzymes. Such efforts are crucial to the development of these therapies that target histone-modifying enzymes within their specific complex. Conclusion Several studies have reported varying degrees of HDACs and HATs expression in many human malignant tissues and there is evidence that substrates of HDACs and HATs are not limited to histones, promoting a cascade of modifications with their deregulation. Considering the exact function of HAT and HDAC is unclear, including the suitable balance between them, future studies may clarify the function of these proteins, improving therapeutic strategies together with existing therapies. Although HDACi have shown efficacy in clinical trials in solid and hematological malignancies, some authors suggest that the use of HDACi should be directed to cases that strongly express HDACs in their tumor cells. Despite these divergences, development and use of HDACs inhibitors are increasing, leading us to continue studying these enzyme expressions and behavior, aiming to determine tumors that will respond better to this type of treatment. Funding This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. Conflict of Interest Statement Authors declare no conflict of interest. References
[1] H. Adams, F.R. Fritzsche, S. Dirnhofer, G. Kristiansen, A. Tzankov, Class I histone deacetylases 1, 2 and 3 are highly expressed in classical Hodgkin's lymphoma, Expert Opin Ther Targets. 14 (2010) 577-584. [2] L.O. Almeida, D.M. Guimaraes, M.D. Martins, M.A.T. Martins, K.A. Warner, J.E. Nor, R.M. Castilho, C.H. Squarize, Unlocking the chromatin of adenoid cystic carcinomas using HDAC inhibitors sensitize cancer stem cells to cisplatin and induces tumor senescence, Stem Cell Res. 21 (2017) 94-105. [3] J.M. Amann, J. Nip, D.K. Strom, B. Lutterbach, H. Harada, N. Lenny, J.R. Downing, S. Meyers, S.W. Hiebert, ETO, a target of t(8;21) in acute leukemia, makes distinct contacts with multiple histone deacetylases and binds mSin3A through its oligomerization domain, Mol Cell Biol. 21 (2001) 64706483. [4] L. Amiri-Kordestani, V. Luchenko, C.J. Peer, K. Ghafourian, J. Reynolds, D. Draper, R. Frye, S. Woo, D. Venzon, J. Wright, M. Skarulis, W.D. Figg, T. Fojo, S.E. Bates, R.L. Piekarz, Phase I trial of a new schedule of romidepsin in patients with advanced cancers, Clin Cancer Res. 19 (2013) 4499-4507. [5] S. Ammanamanchi, M.G. Brattain, Restoration of transforming growth factor-beta signaling through receptor RI induction by histone deacetylase activity inhibition in breast cancer cells, J Biol Chem. 279 (2004) 32620-32625. [6] S. Awasthi, A. Sharma, K. Wong, J. Zhang, E.F. Matlock, L. Rogers, P. Motloch, S. Takemoto, H. Taguchi, M.D. Cole, B. Luscher, O. Dittrich, H. Tagami, Y. Nakatani, M. McGee, A.M. Girard, L. Gaughan, C.N. Robson, R.J. Monnat, Jr., R. Harrod, A human T-cell lymphotropic virus type 1 enhancer of Myc transforming potential stabilizes Myc-TIP60 transcriptional interactions, Mol Cell Biol. 25 (2005) 6178-6198. [7] M.H. Baek, J.Y. Park, C.C. Rhim, Y. Park, K.R. Kim, J.H. Kim, J.H. Nam, Immunohistochemical Characterization of Histone Deacetylase as a Potential Prognostic Marker and Therapeutic Target in Endometrial Stromal Sarcoma, Anticancer Res. 36 (2016) 2527-2534.
[8] D. Bandyopadhyay, N.A. Okan, E. Bales, L. Nascimento, P.A. Cole, E.E. Medrano, Down-regulation of p300/CBP histone acetyltransferase activates a senescence checkpoint in human melanocytes, Cancer Res. 62 (2002) 6231-6239. [9] A.P. Bird, A.P. Wolffe, Methylation-induced repression--belts, braces, and chromatin, Cell. 99 (1999) 451-454. [10] J.V. Bokhman, Two pathogenetic types of endometrial carcinoma, Gynecol Oncol. 15 (1983) 1017. [11] A. Brehm, E.A. Miska, D.J. McCance, J.L. Reid, A.J. Bannister, T. Kouzarides, Retinoblastoma protein recruits histone deacetylase to repress transcription, Nature. 391 (1998) 597-601. [12] M.J. Carrozza, R.T. Utley, J.L. Workman, J. Cote, The diverse functions of histone acetyltransferase complexes, Trends Genet. 19 (2003) 321-329. [13] A. Cashen, M. Juckett, A. Jumonville, M. Litzow, P.J. Flynn, J. Eckardt, B. LaPlant, K. Laumann, C. Erlichman, J. DiPersio, Phase II study of the histone deacetylase inhibitor belinostat (PXD101) for the treatment of myelodysplastic syndrome (MDS), Ann Hematol. 91 (2012) 33-38. [14] C. Caslini, C.D. Capo-chichi, I.H. Roland, E. Nicolas, A.T. Yeung, X.X. Xu, Histone modifications silence the GATA transcription factor genes in ovarian cancer, Oncogene. 25 (2006) 5446-5461. [15] H.H. Chang, C.P. Chiang, H.C. Hung, C.Y. Lin, Y.T. Deng, M.Y. Kuo, Histone deacetylase 2 expression predicts poorer prognosis in oral cancer patients. Oral Oncol, Jul ed., (2009), pp. 610-614. [16] L. Chen, T. Wei, X. Si, Q. Wang, Y. Li, Y. Leng, A. Deng, J. Chen, G. Wang, S. Zhu, J. Kang, Lysine acetyltransferase GCN5 potentiates the growth of non-small cell lung cancer via promotion of E2F1, cyclin D1, and cyclin E1 expression, J Biol Chem. 288 (2013) 14510-14521. [17] X. Cheng, R.M. Blumenthal, Mammalian DNA methyltransferases: a structural perspective, Structure. 16 (2008) 341-350. [18] J.H. Choi, H.J. Kwon, B.I. Yoon, J.H. Kim, S.U. Han, H.J. Joo, D.Y. Kim, Expression profile of histone deacetylase 1 in gastric cancer tissues, Jpn J Cancer Res. 92 (2001) 1300-1304. [19] E.S. Chrun, F. Modolo, D. Vieira, A. Borges-Junior, R.G. Castro, F.I. Daniel, Immunoexpression of HDAC1, HDAC2, and HAT1 in actinic cheilitis and lip squamous cell carcinoma, Oral Dis. (2017). [20] B. Coiffier, B. Pro, H.M. Prince, F. Foss, L. Sokol, M. Greenwood, D. Caballero, P. Borchmann, F. Morschhauser, M. Wilhelm, L. Pinter-Brown, S. Padmanabhan, A. Shustov, J. Nichols, S. Carroll, J. Balser, B. Balser, S. Horwitz, Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T-cell lymphoma after prior systemic therapy, J Clin Oncol. 30 (2012) 631-636. [21] R. Contzler, A. Regamey, B. Favre, T. Roger, D. Hohl, M. Huber, Histone acetyltransferase HBO1 inhibits NF-kappaB activity by coactivator sequestration, Biochem Biophys Res Commun. 350 (2006) 208-213. [22] W.D. Cress, E. Seto, Histone deacetylases, transcriptional control, and cancer, J Cell Physiol. 184 (2000) 1-16. [23] F.I. Daniel, K. Cherubini, L.S. Yurgel, M.A. de Figueiredo, F.G. Salum, The role of epigenetic transcription repression and DNA methyltransferases in cancer, Cancer. 117 (2011) 677-687. [24] A.J. de Ruijter, A.H. van Gennip, H.N. Caron, S. Kemp, A.B. van Kuilenburg, Histone deacetylases (HDACs): characterization of the classical HDAC family, Biochem J. 370 (2003) 737-749. [25] F.J. Dekker, H.J. Haisma, Histone acetyl transferases as emerging drug targets. Drug Discov Today, England, (2009), pp. 942-948. [26] D.S. Dizon, J.A. Blessing, R.T. Penson, R.D. Drake, J.L. Walker, C.M. Johnston, P.A. Disilvestro, A.N. Fader, A phase II evaluation of belinostat and carboplatin in the treatment of recurrent or persistent platinum-resistant ovarian, fallopian tube, or primary peritoneal carcinoma: a Gynecologic Oncology Group study, Gynecol Oncol. 125 (2012) 367-371. [27] D.S. Dizon, L. Damstrup, N.J. Finkler, U. Lassen, P. Celano, R. Glasspool, E. Crowley, H.S. Lichenstein, P. Knoblach, R.T. Penson, Phase II activity of belinostat (PXD-101), carboplatin, and paclitaxel in women with previously treated ovarian cancer, Int J Gynecol Cancer. 22 (2012) 979-986.
[28] M. Dokmanovic, P.A. Marks, Prospects: histone deacetylase inhibitors, J Cell Biochem. 96 (2005) 293-304. [29] A.P. Feinberg, B. Tycko, The history of cancer epigenetics, Nat Rev Cancer. 4 (2004) 143-153. [30] L. Feng, M. Pan, J. Sun, H. Lu, Q. Shen, S. Zhang, T. Jiang, L. Liu, W. Jin, Y. Chen, X. Wang, H. Jin, Histone deacetylase 3 inhibits expression of PUMA in gastric cancer cells, J Mol Med (Berl). 91 (2013) 49-58. [31] L. Gaughan, I.R. Logan, S. Cook, D.E. Neal, C.N. Robson, Tip60 and histone deacetylase 1 regulate androgen receptor activity through changes to the acetylation status of the receptor, J Biol Chem. 277 (2002) 25904-25913. [32] S.A. Gayther, S.J. Batley, L. Linger, A. Bannister, K. Thorpe, S.F. Chin, Y. Daigo, P. Russell, A. Wilson, H.M. Sowter, J.D. Delhanty, B.A. Ponder, T. Kouzarides, C. Caldas, Mutations truncating the EP300 acetylase in human cancers, Nat Genet. 24 (2000) 300-303. [33] G. Giaccone, A. Rajan, A. Berman, R.J. Kelly, E. Szabo, A. Lopez-Chavez, J. Trepel, M.J. Lee, L. Cao, I. Espinoza-Delgado, J. Spittler, P.J. Loehrer, Sr., Phase II study of belinostat in patients with recurrent or refractory advanced thymic epithelial tumors, J Clin Oncol. 29 (2011) 2052-2059. [34] C. Giaginis, P. Alexandrou, I. Delladetsima, I. Giannopoulou, E. Patsouris, S. Theocharis, Clinical significance of histone deacetylase (HDAC)-1, HDAC-2, HDAC-4, and HDAC-6 expression in human malignant and benign thyroid lesions, Tumour Biol. 35 (2014) 61-71. [35] M.A. Glozak, E. Seto, Histone deacetylases and cancer, Oncogene. 26 (2007) 5420-5432. [36] M. Gotte, MicroRNAs in breast cancer pathogenesis, Minerva Ginecol. 62 (2010) 559-571. [37] P.A. Grant, A. Eberharter, S. John, R.G. Cook, B.M. Turner, J.L. Workman, Expanded lysine acetylation specificity of Gcn5 in native complexes, J Biol Chem. 274 (1999) 5895-5900. [38] I.V. Gregoretti, Y.M. Lee, H.V. Goodson, Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis, J Mol Biol. 338 (2004) 17-31. [39] M. Grigat, Y. Jaschke, F. Kliewe, M. Pfeifer, S. Walz, H.J. Schuller, Multiple histone deacetylases are recruited by corepressor Sin3 and contribute to gene repression mediated by Opi1 regulator of phospholipid biosynthesis in the yeast Saccharomyces cerevisiae, Mol Genet Genomics. 287 (2012) 461-472. [40] B. Gruhn, T. Naumann, D. Gruner, M. Walther, S. Wittig, S. Becker, J.F. Beck, J. Sonnemann, The expression of histone deacetylase 4 is associated with prednisone poor-response in childhood acute lymphoblastic leukemia, Leuk Res. 37 (2013) 1200-1207. [41] M. Haberland, R.L. Montgomery, E.N. Olson, The many roles of histone deacetylases in development and physiology: implications for disease and therapy, Nat Rev Genet. 10 (2009) 32-42. [42] K. Halkidou, L. Gaughan, S. Cook, H.Y. Leung, D.E. Neal, C.N. Robson, Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer, Prostate. 59 (2004) 177-189. [43] K. Halkidou, V.J. Gnanapragasam, P.B. Mehta, I.R. Logan, M.E. Brady, S. Cook, H.Y. Leung, D.E. Neal, C.N. Robson, Expression of Tip60, an androgen receptor coactivator, and its role in prostate cancer development, Oncogene. 22 (2003) 2466-2477. [44] C.A. Hassig, T.C. Fleischer, A.N. Billin, S.L. Schreiber, D.E. Ayer, Histone deacetylase activity is required for full transcriptional repression by mSin3A. Cell, United States, (1997), pp. 341-347. [45] A. Hayashi, A. Horiuchi, N. Kikuchi, T. Hayashi, C. Fuseya, A. Suzuki, I. Konishi, T. Shiozawa, Typespecific roles of histone deacetylase (HDAC) overexpression in ovarian carcinoma: HDAC1 enhances cell proliferation and HDAC3 stimulates cell migration with downregulation of E-cadherin, Int J Cancer. 127 (2010) 1332-1346. [46] C.A. Hobbs, G. Wei, K. DeFeo, B. Paul, C.S. Hayes, S.K. Gilmour, Tip60 protein isoforms and altered function in skin and tumors that overexpress ornithine decarboxylase, Cancer Res. 66 (2006) 8116-8122. [47] A. Hrzenjak, F. Moinfar, M.L. Kremser, B. Strohmeier, P.B. Staber, K. Zatloukal, H. Denk, Valproate inhibition of histone deacetylase 2 affects differentiation and decreases proliferation of endometrial stromal sarcoma cells, Mol Cancer Ther. 5 (2006) 2203-2210.
[48] C. Hu, M. Liu, W. Zhang, Q. Xu, K. Ma, L. Chen, Z. Wang, S. He, H. Zhu, N. Xu, Upregulation of KLF4 by methylseleninic acid in human esophageal squamous cell carcinoma cells: Modification of histone H3 acetylation through HAT/HDAC interplay, Mol Carcinog. (2014). [49] J. Hu, C. Bernatchez, L. Zhang, X. Xia, E.S. Kleinerman, M.C. Hung, P. Hwu, S. Li, Induction of NKG2D Ligands on Solid Tumors Requires Tumor-Specific CD8+ T Cells and Histone Acetyltransferases, Cancer Immunol Res. 5 (2017) 300-311. [50] B.H. Huang, M. Laban, C.H. Leung, L. Lee, C.K. Lee, M. Salto-Tellez, G.C. Raju, S.C. Hooi, Inhibition of histone deacetylase 2 increases apoptosis and p21Cip1/WAF1 expression, independent of histone deacetylase 1, Cell Death Differ. 12 (2005) 395-404. [51] K. Ishihama, M. Yamakawa, S. Semba, H. Takeda, S. Kawata, S. Kimura, W. Kimura, Expression of HDAC1 and CBP/p300 in human colorectal carcinomas, J Clin Pathol. 60 (2007) 1205-1210. [52] N.G. Iyer, J. Xian, S.F. Chin, A.J. Bannister, Y. Daigo, S. Aparicio, T. Kouzarides, C. Caldas, p300 is required for orderly G1/S transition in human cancer cells, Oncogene. 26 (2007) 21-29. [53] A. Izzo, K. Kamieniarz, R. Schneider, The histone H1 family: specific members, specific functions?, Biol Chem. 389 (2008) 333-343. [54] A. Jarzab, J.J. Luszczki, M. Guz, E. Gumbarewicz, K. Polberg, A. Stepulak, Additive Interaction of Cisplatin and Histone Deacetylase Inhibitors Combined Treatment in Rhabdomyosarcoma Cells - An Isobolographic Analysis, Anticancer Res. 37 (2017) 1067-1074. [55] B.M. Javierre, A.F. Fernandez, J. Richter, F. Al-Shahrour, J.I. Martin-Subero, J. Rodriguez-Ubreva, M. Berdasco, M.F. Fraga, T.P. O'Hanlon, L.G. Rider, F.V. Jacinto, F.J. Lopez-Longo, J. Dopazo, M. Forn, M.A. Peinado, L. Carreno, A.H. Sawalha, J.B. Harley, R. Siebert, M. Esteller, F.W. Miller, E. Ballestar, Changes in the pattern of DNA methylation associate with twin discordance in systemic lupus erythematosus, Genome Res. 20 (2010) 170-179. [56] S.F. Jones, J.R. Infante, D.R. Spigel, N.W. Peacock, D.S. Thompson, F.A. Greco, W. McCulloch, H.A. Burris, 3rd, Phase 1 results from a study of romidepsin in combination with gemcitabine in patients with advanced solid tumors, Cancer Invest. 30 (2012) 481-486. [57] L.J. Juan, W.J. Shia, M.H. Chen, W.M. Yang, E. Seto, Y.S. Lin, C.W. Wu, Histone deacetylases specifically down-regulate p53-dependent gene activation, J Biol Chem. 275 (2000) 20436-20443. [58] R. Kanwal, S. Gupta, Epigenetic modifications in cancer, Clinical Genetics. 81 (2014) 303-311. [59] E. Karouzakis, R.E. Gay, S. Gay, M. Neidhart, Epigenetic control in rheumatoid arthritis synovial fibroblasts, Nat Rev Rheumatol. 5 (2009) 266-272. [60] S. Karthik, R. Sankar, K. Varunkumar, V. Ravikumar, Romidepsin induces cell cycle arrest, apoptosis, histone hyperacetylation and reduces matrix metalloproteinases 2 and 9 expression in bortezomib sensitized non-small cell lung cancer cells, Biomed Pharmacother. 68 (2014) 327-334. [61] T. Katsumoto, Y. Aikawa, A. Iwama, S. Ueda, H. Ichikawa, T. Ochiya, I. Kitabayashi, MOZ is essential for maintenance of hematopoietic stem cells, Genes Dev. 20 (2006) 1321-1330. [62] W.K. Kelly, O.A. O'Connor, L.M. Krug, J.H. Chiao, M. Heaney, T. Curley, B. MacGregore-Cortelli, W. Tong, J.P. Secrist, L. Schwartz, S. Richardson, E. Chu, S. Olgac, P.A. Marks, H. Scher, V.M. Richon, Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer, J Clin Oncol. 23 (2005) 3923-3931. [63] O. Khan, N.B. La Thangue, HDAC inhibitors in cancer biology: emerging mechanisms and clinical applications, Immunol Cell Biol. 90 (2012) 85-94. [64] S.M. Khoshnaw, A.R. Green, D.G. Powe, I.O. Ellis, MicroRNA involvement in the pathogenesis and management of breast cancer, J Clin Pathol. 62 (2009) 422-428. [65] J.H. Kim, B. Kim, L. Cai, H.J. Choi, K.A. Ohgi, C. Tran, C. Chen, C.H. Chung, O. Huber, D.W. Rose, C.L. Sawyers, M.G. Rosenfeld, S.H. Baek, Transcriptional regulation of a metastasis suppressor gene by Tip60 and beta-catenin complexes, Nature. 434 (2005) 921-926. [66] M.S. Kim, H.J. Kwon, Y.M. Lee, J.H. Baek, J.E. Jang, S.W. Lee, E.J. Moon, H.S. Kim, S.K. Lee, H.Y. Chung, C.W. Kim, K.W. Kim, Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes, Nat Med. 7 (2001) 437-443.
[67] M.S. Kim, H.J. Kwon, Y.M. Lee, J.H. Baek, J.E. Jang, S.W. Lee, E.J. Moon, H.S. Kim, S.K. Lee, H.Y. Chung, C.W. Kim, K.W. Kim, Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat Med, United States, (2001), pp. 437-443. [68] M.H. Kirschbaum, K.A. Foon, P. Frankel, C. Ruel, B. Pulone, J.M. Tuscano, E.M. Newman, A phase 2 study of belinostat (PXD101) in patients with relapsed or refractory acute myeloid leukemia or patients over the age of 60 with newly diagnosed acute myeloid leukemia: a California Cancer Consortium Study, Leuk Lymphoma. 55 (2014) 2301-2304. [69] E. Klieser, R. Urbas, S. Stattner, F. Primavesi, T. Jager, A. Dinnewitzer, C. Mayr, T. Kiesslich, K. Holzmann, P. Di Fazio, D. Neureiter, S. Swierczynski, Comprehensive immunohistochemical analysis of histone deacetylases in pancreatic neuroendocrine tumors: HDAC5 as a predictor of poor clinical outcome, Hum Pathol. (2017). [70] L.R. Kong, T.Z. Tan, W.R. Ong, C. Bi, H. Huynh, S.C. Lee, W.J. Chng, P.J. Eichhorn, B.C. Goh, Belinostat exerts anti-tumor cytotoxicity through the ubiquitin-proteasome pathway in lung squamous cell carcinoma, Mol Oncol. (2017). [71] A.R. Krebs, K. Karmodiya, M. Lindahl-Allen, K. Struhl, L. Tora, SAGA and ATAC histone acetyl transferase complexes regulate distinct sets of genes and ATAC defines a class of p300-independent enhancers, Mol Cell. 44 (2011) 410-423. [72] C.A. Krusche, A.J. Vloet, I. Classen-Linke, U. von Rango, H.M. Beier, J. Alfer, Class I histone deacetylase expression in the human cyclic endometrium and endometrial adenocarcinomas. Hum Reprod, England, (2007), 2956-2966. [73] C.A. Krusche, P. Wulfing, C. Kersting, A. Vloet, W. Bocker, L. Kiesel, H.M. Beier, J. Alfer, Histone deacetylase-1 and -3 protein expression in human breast cancer: a tissue microarray analysis, Breast Cancer Res Treat. 90 (2005) 15-23. [74] V. Kumar, A.K. Abbas, J.C. Aster, Roobins Basic Pathology, 9 ed., 2012. [75] C.H. Kuo, C.C. Hsieh, M.S. Lee, K.T. Chang, H.F. Kuo, C.H. Hung, Epigenetic regulation in allergic diseases and related studies, Asia Pac Allergy. 4 (2014) 14-18. [76] C.D. Laherty, W.M. Yang, J.M. Sun, J.R. Davie, E. Seto, R.N. Eisenman, Histone deacetylases associated with the mSin3 corepressor mediate mad transcriptional repression, Cell. 89 (1997) 349356. [77] K.C. Lakshmaiah, L.A. Jacob, S. Aparna, D. Lokanatha, S.C. Saldanha, Epigenetic therapy of cancer with histone deacetylase inhibitors, J Cancer Res Ther. 10 (2014) 469-478. [78] A.A. Lane, B.A. Chabner, Histone deacetylase inhibitors in cancer therapy, J Clin Oncol. 27 (2009) 5459-5468. [79] S.F. Lax, Molecular genetic pathways in various types of endometrial carcinoma: from a phenotypical to a molecular-based classification, Virchows Arch. 444 (2004) 213-223. [80] C. Leduc, P. Claverie, B. Eymin, E. Col, S. Khochbin, E. Brambilla, S. Gazzeri, p14ARF promotes RB accumulation through inhibition of its Tip60-dependent acetylation, Oncogene. 25 (2006) 41474154. [81] S.H. Lee, C. Yoo, S. Im, J.H. Jung, H.J. Choi, J. Yoo, Expression of Histone Deacetylases in Diffuse Large B-cell Lymphoma and Its Clinical Significance, Int J Med Sci. 11 (2014) 994-1000. [82] Y.C. Lin, J.H. Lin, C.W. Chou, Y.F. Chang, S.H. Yeh, C.C. Chen, Statins increase p21 through inhibition of histone deacetylase activity and release of promoter-associated HDAC1/2, Cancer Res. 68 (2008) 2375-2383. [83] S. Liu, L. Shi, X. Yang, D. Ye, T. Wang, C. Dong, W. Guo, Y. Liao, H. Song, D. Xu, J. Hu, Z. Zhang, J. Deng, Nuclear survivin promoted by acetylation is associated with the aggressive phenotype of oral squamous cell carcinoma, Cell Cycle. (2017) 1-9. [84] R.X. Luo, A.A. Postigo, D.C. Dean, Rb interacts with histone deacetylase to repress transcription, Cell. 92 (1998) 463-473. [85] B.B. Ma, F. Sung, Q. Tao, F.F. Poon, V.W. Lui, W. Yeo, S.L. Chan, A.T. Chan, The preclinical activity of the histone deacetylase inhibitor PXD101 (belinostat) in hepatocellular carcinoma cell lines, Invest New Drugs. 28 (2010) 107-114.
[86] T. Ma, F. Galimberti, C.P. Erkmen, V. Memoli, F. Chinyengetere, L. Sempere, J.H. Beumer, B.N. Anyang, W. Nugent, D. Johnstone, G.J. Tsongalis, J.M. Kurie, H. Li, J. Direnzo, Y. Guo, S.J. Freemantle, K.H. Dragnev, E. Dmitrovsky, Comparing histone deacetylase inhibitor responses in genetically engineered mouse lung cancer models and a window of opportunity trial in patients with lung cancer, Mol Cancer Ther. 12 (2013) 1545-1555. [87] X. Ma, J. Wang, J. Liu, Q. Mo, X. Yan, D. Ma, H. Duan, Targeting CD146 in combination with vorinostat for the treatment of ovarian cancer cells, Oncol Lett. 13 (2017) 1681-1687. [88] Y. Ma, Y. Yue, M. Pan, J. Sun, J. Chu, X. Lin, W. Xu, L. Feng, Y. Chen, D. Chen, V.Y. Shin, X. Wang, H. Jin, Histone deacetylase 3 inhibits new tumor suppressor gene DTWD1 in gastric cancer, Am J Cancer Res. 5 (2015) 663-673. [89] H.J. Mackay, H. Hirte, T. Colgan, A. Covens, K. MacAlpine, P. Grenci, L. Wang, J. Mason, P.A. Pham, M.S. Tsao, J. Pan, J. Zwiebel, A.M. Oza, Phase II trial of the histone deacetylase inhibitor belinostat in women with platinum resistant epithelial ovarian cancer and micropapillary (LMP) ovarian tumours, Eur J Cancer. 46 (2010) 1573-1579. [90] B.S. Mann, J.R. Johnson, M.H. Cohen, R. Justice, R. Pazdur, FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma, Oncologist. 12 (2007) 1247-1252. [91] P. Marks, R.A. Rifkind, V.M. Richon, R. Breslow, T. Miller, W.K. Kelly, Histone deacetylases and cancer: causes and therapies, Nat Rev Cancer. 1 (2001) 194-202. [92] C. Meisenberg, M.E. Ashour, L. El-Shafie, C. Liao, A. Hodgson, A. Pilborough, S.A. Khurram, J.A. Downs, S.E. Ward, S.F. El-Khamisy, Epigenetic changes in histone acetylation underpin resistance to the topoisomerase I inhibitor irinotecan, Nucleic Acids Res. (2016). [93] F. Miao, D.D. Smith, L. Zhang, A. Min, W. Feng, R. Natarajan, Lymphocytes from patients with type 1 diabetes display a distinct profile of chromatin histone H3 lysine 9 dimethylation: an epigenetic study in diabetes, Diabetes. 57 (2008) 3189-3198. [94] S.K. Min, Y.H. Koh, Y. Park, H.J. Kim, J. Seo, H.R. Park, S.J. Cho, I.S. Kim, Expression of HAT1 and HDAC1, 2, 3 in Diffuse Large B-Cell Lymphomas, Peripheral T-Cell Lymphomas, and NK/T-Cell Lymphomas, Korean J Pathol. 46 (2012) 142-150. [95] F. Moraga, F. Aquea, Composition of the SAGA complex in plants and its role in controlling gene expression in response to abiotic stresses, Front Plant Sci. 6 (2015) 865. [96] D.A. Moreno, C.A. Scrideli, M.A. Cortez, R. de Paula Queiroz, E.T. Valera, V. da Silva Silveira, J.A. Yunes, S.R. Brandalise, L.G. Tone, Differential expression of HDAC3, HDAC7 and HDAC9 is associated with prognosis and survival in childhood acute lymphoblastic leukaemia, Br J Haematol. 150 (2010) 665-673. [97] M. Mottamal, S. Zheng, T.L. Huang, G. Wang, Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents, Molecules. 20 (2015) 3898-3941. [98] B.M. Muller, L. Jana, A. Kasajima, A. Lehmann, J. Prinzler, J. Budczies, K.J. Winzer, M. Dietel, W. Weichert, C. Denkert, Differential expression of histone deacetylases HDAC1, 2 and 3 in human breast cancer--overexpression of HDAC2 and HDAC3 is associated with clinicopathological indicators of disease progression, BMC Cancer. 13 (2013) 215. [99] M.C. Myzak, W.M. Dashwood, G.A. Orner, E. Ho, R.H. Dashwood, Sulforaphane inhibits histone deacetylase in vivo and suppresses tumorigenesis in Apc-minus mice, Faseb j. 20 (2006) 506-508. [100] M. Nakagawa, Y. Oda, T. Eguchi, S. Aishima, T. Yao, F. Hosoi, Y. Basaki, M. Ono, M. Kuwano, M. Tanaka, M. Tsuneyoshi, Expression profile of class I histone deacetylases in human cancer tissues, Oncol Rep. 18 (2007) 769-774. [101] C. Nanavati, D.E. Mager, Sequential Exposure of Bortezomib and Vorinostat is Synergistic in Multiple Myeloma Cells, Pharm Res. 34 (2017) 668-679. [102] H.H. Ng, A. Bird, DNA methylation and chromatin modification, Curr Opin Genet Dev. 9 (1999) 158-163. [103] H. Ogiwara, M. Sasaki, T. Mitachi, T. Oike, S. Higuchi, Y. Tominaga, T. Kohno, Targeting p300 addiction in CBP-deficient cancers causes synthetic lethality via apoptotic cell death due to abrogation of MYC expression, Cancer Discov. (2015).
[104] K. Ohta, M. Ohigashi, A. Naganawa, H. Ikeda, M. Sakai, J. Nishikawa, M. Imagawa, S. Osada, T. Nishihara, Histone acetyltransferase MOZ acts as a co-activator of Nrf2-MafK and induces tumour marker gene expression during hepatocarcinogenesis, Biochem J. 402 (2007) 559-566. [105] S.K. Patra, A. Patra, R. Dahiya, Histone deacetylase and DNA methyltransferase in human prostate cancer, Biochem Biophys Res Commun. 287 (2001) 705-713. [106] C.L. Peterson, M.A. Laniel, Histones and histone modifications, Curr Biol. 14 (2004) R546-551. [107] D.K. Phi van, E. Muhlbauer, L. Phi-van, Histone deacetylase HDAC1 downregulates transcription of the serotonin transporter (5-HTT) gene in tumor cells, Biochim Biophys Acta. 1849 (2015) 909918. [108] R.L. Piekarz, R. Frye, M. Turner, J.J. Wright, S.L. Allen, M.H. Kirschbaum, J. Zain, H.M. Prince, J.P. Leonard, L.J. Geskin, C. Reeder, D. Joske, W.D. Figg, E.R. Gardner, S.M. Steinberg, E.S. Jaffe, M. Stetler-Stevenson, S. Lade, A.T. Fojo, S.E. Bates, Phase II multi-institutional trial of the histone deacetylase inhibitor romidepsin as monotherapy for patients with cutaneous T-cell lymphoma, J Clin Oncol. 27 (2009) 5410-5417. [109] R. Pili, G. Liu, S. Chintala, H. Verheul, S. Rehman, K. Attwood, M.A. Lodge, R. Wahl, J.I. Martin, K.M. Miles, S. Paesante, R. Adelaiye, A. Godoy, S. King, J. Zwiebel, M.A. Carducci, Combination of the histone deacetylase inhibitor vorinostat with bevacizumab in patients with clear-cell renal cell carcinoma: a multicentre, single-arm phase I/II clinical trial, Br J Cancer. 116 (2017) 874-883. [110] R.M. Poole, Belinostat: first global approval, Drugs. 74 (2014) 1543-1554. [111] A. Portela, M. Esteller, Epigenetic modifications and human disease, Nat Biotechnol. 28 (2010) 1057-1068. [112] M. Qian, H. Zhang, S.K. Kham, S. Liu, C. Jiang, X. Zhao, Y. Lu, C. Goodings, T.N. Lin, R. Zhang, T. Moriyama, Z. Yin, Z. Li, T.C. Quah, H. Ariffin, A.M. Tan, S. Shen, D. Bhojwani, S. Hu, S. Chen, H. Zheng, C.H. Pui, A.E. Yeoh, J.J. Yang, Whole-transcriptome sequencing identifies a distinct subtype of acute lymphoblastic leukemia with predominant genomic abnormalities of EP300 and CREBBP, Genome Res. 27 (2017) 185-195. [113] S.S. Ramalingam, C.P. Belani, C. Ruel, P. Frankel, B. Gitlitz, M. Koczywas, I. Espinoza-Delgado, D. Gandara, Phase II study of belinostat (PXD101), a histone deacetylase inhibitor, for second line therapy of advanced malignant pleural mesothelioma, J Thorac Oncol. 4 (2009) 97-101. [114] K.C. Ravindra, B.R. Selvi, M. Arif, B.A.A. Reddy, G.R. Thanuja, S. Agrawal, S.K. Pradhan, N. Nagashayana, D. Dasgupta, T.K. Kundu, Inhibition of Lysine Acetyltransferase KAT3B/p300 Activity by a Naturally Occurring Hydroxynaphthoquinone, Plumbagin*. J Biol Chem, (2009), pp. 24453-24464. [115] A. Riss, E. Scheer, M. Joint, S. Trowitzsch, I. Berger, L. Tora, Subunits of ADA-Two-A-Containing (ATAC) or Spt-Ada-Gcn5-Acetyltrasferase (SAGA) Coactivator Complexes Enhance the Acetyltransferase Activity of GCN5, J Biol Chem. (2015). [116] F.M. Robertson, K. Chu, K.M. Boley, Z. Ye, H. Liu, M.C. Wright, R. Moraes, X. Zhang, T.L. Green, S.H. Barsky, C. Heise, M. Cristofanilli, The class I HDAC inhibitor Romidepsin targets inflammatory breast cancer tumor emboli and synergizes with paclitaxel to inhibit metastasis, J Exp Ther Oncol. 10 (2013) 219-233. [117] S.Y. Roth, J.M. Denu, C.D. Allis, Histone acetyltransferases, Annu Rev Biochem. 70 (2001) 81120. [118] M.G. Saelen, A.H. Ree, A. Kristian, K.G. Fleten, T. Furre, H.H. Hektoen, K. Flatmark, Radiosensitization by the histone deacetylase inhibitor vorinostat under hypoxia and with capecitabine in experimental colorectal carcinoma, Radiat Oncol. 7 (2012) 165. [119] T. Sakuma, K. Uzawa, T. Onda, M. Shiiba, H. Yokoe, T. Shibahara, H. Tanzawa, Aberrant expression of histone deacetylase 6 in oral squamous cell carcinoma, Int J Oncol. 29 (2006) 117-124. [120] R. Sarfstein, I. Bruchim, A. Fishman, H. Werner, The mechanism of action of the histone deacetylase inhibitor vorinostat involves interaction with the insulin-like growth factor signaling pathway, PLoS One. 6 (2011) e24468. [121] H. Sasaki, S. Moriyama, Y. Nakashima, Y. Kobayashi, M. Kiriyama, I. Fukai, Y. Yamakawa, Y. Fujii, Histone deacetylase 1 mRNA expression in lung cancer, Lung Cancer. 46 (2004) 171-178.
[122] J. Savickiene, G. Treigyte, G. Valiuliene, I. Stirblyte, R. Navakauskiene, Epigenetic and molecular mechanisms underlying the antileukemic activity of the histone deacetylase inhibitor belinostat in human acute promyelocytic leukemia cells, Anticancer Drugs. 25 (2014) 938-949. [123] S. Scheipl, B. Lohberger, B. Rinner, E.V. Froehlich, A. Beham, F. Quehenberger, A. Lazary, P. Pal Varga, J. Haybaeck, A. Leithner, B. Liegl, Histone deacetylase inhibitors as potential therapeutic approaches for chordoma: an immunohistochemical and functional analysis, J Orthop Res. 31 (2013) 1999-2005. [124] D.B. Seligson, S. Horvath, T. Shi, H. Yu, S. Tze, M. Grunstein, S.K. Kurdistani, Global histone modification patterns predict risk of prostate cancer recurrence, Nature. 435 (2005) 1262-1266. [125] J. Seo, S.K. Min, H.R. Park, D.H. Kim, M.J. Kwon, L.S. Kim, Y.S. Ju, Expression of Histone Deacetylases HDAC1, HDAC2, HDAC3, and HDAC6 in Invasive Ductal Carcinomas of the Breast, J Breast Cancer. 17 (2014) 323-331. [126] M. Sharma, M. Zarnegar, X. Li, B. Lim, Z. Sun, Androgen receptor interacts with a novel MYST protein, HBO1, J Biol Chem. 275 (2000) 35200-35208. [127] M. Shi, X.J. Lu, J. Zhang, H. Diao, G. Li, L. Xu, T. Wang, J. Wei, W. Meng, J.L. Ma, H. Yu, Y.G. Wang, Oridonin, a novel lysine acetyltransferases inhibitor, inhibits proliferation and induces apoptosis in gastric cancer cells through p53- and caspase-3-mediated mechanisms, Oncotarget. 7 (2016) 22623-22631. [128] W. Shi, Y.R. Lawrence, H. Choy, M. Werner-Wasik, D.W. Andrews, J.J. Evans, K.D. Judy, C.J. Farrell, Y. Moshel, A.C. Berger, V. Bar-Ad, A.P. Dicker, Vorinostat as a radiosensitizer for brain metastasis: a phase I clinical trial, J Neurooncol. 118 (2014) 313-319. [129] A. Shustov, B. Coiffier, S. Horwitz, L. Sokol, B. Pro, J. Wolfson, B. Balser, R. Eisch, L. Popplewell, H.M. Prince, S.L. Allen, R. Piekarz, S. Bates, Romidepsin is effective and well tolerated in older patients with peripheral T-cell lymphoma: analysis of two phase II trials, Leuk Lymphoma. (2017) 1-7. [130] P.J. Skene, S. Henikoff, Histone variants in pluripotency and disease, Development. 140 (2013) 2513-2524. [131] J. Song, J.H. Noh, J.H. Lee, J.W. Eun, Y.M. Ahn, S.Y. Kim, S.H. Lee, W.S. Park, N.J. Yoo, J.Y. Lee, S.W. Nam, Increased expression of histone deacetylase 2 is found in human gastric cancer, Apmis. 113 (2005) 264-268. [132] B.D. Strahl, C.D. Allis, The language of covalent histone modifications, Nature. 403 (2000) 4145. [133] W.J. Sun, H. Huang, B. He, D.H. Hu, P.H. Li, Y.J. Yu, X.H. Zhou, Z. Lv, L. Zhou, T.Y. Hu, Z.C. Yao, M.D. Lu, X. Shen, Z.Q. Zheng, Romidepsin induces G2/M phase arrest via Erk/cdc25C/cdc2/cyclinB pathway and apoptosis induction through JNK/c-Jun/caspase3 pathway in hepatocellular carcinoma cells, Biochem Pharmacol. 127 (2017) 90-100. [134] J. Suzuki, Y.Y. Chen, G.K. Scott, S. Devries, K. Chin, C.C. Benz, F.M. Waldman, E.S. Hwang, Protein acetylation and histone deacetylase expression associated with malignant breast cancer progression, Clin Cancer Res. 15 (2009) 3163-3171. [135] S. Theocharis, J. Klijanienko, C. Giaginis, J. Rodriguez, T. Jouffroy, A. Girod, P. Alexandrou, X. Sastre-Garau, Histone deacetylase-1 and -2 expression in mobile tongue squamous cell carcinoma: associations with clinicopathological parameters and patients survival, J Oral Pathol Med. 40 (2011) 706-714. [136] A. Thomas, A. Rajan, E. Szabo, Y. Tomita, C.A. Carter, B. Scepura, A. Lopez-Chavez, M.J. Lee, C.E. Redon, A. Frosch, C.J. Peer, Y. Chen, R. Piekarz, S.M. Steinberg, J.B. Trepel, W.D. Figg, D.S. Schrump, G. Giaccone, A phase I/II trial of belinostat in combination with cisplatin, doxorubicin, and cyclophosphamide in thymic epithelial tumors: a clinical and translational study, Clin Cancer Res. 20 (2014) 5392-5402. [137] K.K. To, W.S. Tong, L.W. Fu, Reversal of platinum drug resistance by the histone deacetylase inhibitor belinostat, Lung Cancer. 103 (2017) 58-65. [138] Z.T. Tong, M.Y. Cai, X.G. Wang, L.L. Kong, S.J. Mai, Y.H. Liu, H.B. Zhang, Y.J. Liao, F. Zheng, W. Zhu, T.H. Liu, X.W. Bian, X.Y. Guan, M.C. Lin, M.S. Zeng, Y.X. Zeng, H.F. Kung, D. Xie, EZH2 supports
nasopharyngeal carcinoma cell aggressiveness by forming a co-repressor complex with HDAC1/HDAC2 and Snail to inhibit E-cadherin, Oncogene. 31 (2012) 583-594. [139] R.G. Urdinguio, J.V. Sanchez-Mut, M. Esteller, Epigenetic mechanisms in neurological diseases: genes, syndromes, and therapies, Lancet Neurol. 8 (2009) 1056-1072. [140] K. Ververis, T.C. Karagiannis, An atlas of histone deacetylase expression in breast cancer: fluorescence methodology for comparative semi-quantitative analysis, Am J Transl Res. 4 (2012) 2443. [141] J. Vitfell-Rasmussen, I. Judson, A. Safwat, R.L. Jones, P.B. Rossen, M. Lind-Hansen, P. Knoblauch, A. Krarup-Hansen, A Phase I/II Clinical Trial of Belinostat (PXD101) in Combination with Doxorubicin in Patients with Soft Tissue Sarcomas, Sarcoma. 2016 (2016). [142] J. von Burstin, S. Eser, M.C. Paul, B. Seidler, M. Brandl, M. Messer, A. von Werder, A. Schmidt, J. Mages, P. Pagel, A. Schnieke, R.M. Schmid, G. Schneider, D. Saur, E-cadherin regulates metastasis of pancreatic cancer in vivo and is suppressed by a SNAIL/HDAC1/HDAC2 repressor complex, Gastroenterology. 137 (2009) 361-371, 371.e361-365. [143] Y.L. Wang, F. Faiola, M. Xu, S. Pan, E. Martinez, Human ATAC Is a GCN5/PCAF-containing acetylase complex with a novel NC2-like histone fold module that interacts with the TATA-binding protein, J Biol Chem. 283 (2008) 33808-33815. [144] R. Warrener, H. Beamish, A. Burgess, N.J. Waterhouse, N. Giles, D. Fairlie, B. Gabrielli, Tumor cell-selective cytotoxicity by targeting cell cycle checkpoints, Faseb j. 17 (2003) 1550-1552. [145] W. Weichert, HDAC expression and clinical prognosis in human malignancies, Cancer Lett. 280 (2009) 168-176. [146] W. Weichert, A. Roske, V. Gekeler, T. Beckers, C. Stephan, K. Jung, F.R. Fritzsche, S. Niesporek, C. Denkert, M. Dietel, G. Kristiansen, Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy, Br J Cancer. 98 (2008) 604-610. [147] S.J. Whittaker, M.F. Demierre, E.J. Kim, A.H. Rook, A. Lerner, M. Duvic, J. Scarisbrick, S. Reddy, T. Robak, J.C. Becker, A. Samtsov, W. McCulloch, Y.H. Kim, Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma, J Clin Oncol. 28 (2010) 4485-4491. [148] N. Yamada, T. Hamada, M. Goto, H. Tsutsumida, M. Higashi, M. Nomoto, S. Yonezawa, MUC2 expression is regulated by histone H3 modification and DNA methylation in pancreatic cancer, Int J Cancer. 119 (2006) 1850-1857. [149] H. Yang, S. Maddipoti, A. Quesada, Z. Bohannan, M. Cabrero Calvo, S. Colla, Y. Wei, M. Estecio, W. Wierda, C. Bueso-Ramos, G. Garcia-Manero, Analysis of class I and II histone deacetylase gene expression in human leukemia, Leuk Lymphoma. (2015) 1-8. [150] C.B. Yoo, P.A. Jones, Epigenetic therapy of cancer: past, present and future, Nat Rev Drug Discov. 5 (2006) 37-50. [151] L. Zhang, M. Cai, Z. Gong, B. Zhang, Y. Li, L. Guan, X. Hou, Q. Li, G. Liu, Z. Xue, M.H. Yang, J. Ye, Y.E. Chin, H. You, Geminin facilitates FoxO3 deacetylation to promote breast cancer cell metastasis, J Clin Invest. (2017). [152] N. Zhang, W. Ichikawa, F. Faiola, S.Y. Lo, X. Liu, E. Martinez, MYC interacts with the human STAGA coactivator complex via multivalent contacts with the GCN5 and TRRAP subunits, Biochim Biophys Acta. 1839 (2014) 395-405. [153] Y. Zhao, S. Lu, L. Wu, G. Chai, H. Wang, Y. Chen, J. Sun, Y. Yu, W. Zhou, Q. Zheng, M. Wu, G.A. Otterson, W.G. Zhu, Acetylation of p53 at lysine 373/382 by the histone deacetylase inhibitor depsipeptide induces expression of p21(Waf1/Cip1), Mol Cell Biol. 26 (2006) 2782-2790. [154] M.I. Zhou, R.L. Foy, V.C. Chitalia, J. Zhao, M.V. Panchenko, H. Wang, H.T. Cohen, Jade-1, a candidate renal tumor suppressor that promotes apoptosis, Proc Natl Acad Sci U S A. 102 (2005) 11035-11040. [155] P. Zhu, E. Martin, J. Mengwasser, P. Schlag, K.P. Janssen, M. Gottlicher, Induction of HDAC2 expression upon loss of APC in colorectal tumorigenesis, Cancer Cell. 5 (2004) 455-463.
FIGURES LEGENDS Fig. 1. HAT and HDAC action. Acetylation catalyzed by HAT affects transcription through neutralization of the positive charge of histones, weakens interactions between DNA and histone or among nucleosomes, thus reducing chromatin compaction and favoring gene transcription. In an opposite way, HDACs promote deacetylation returning it to chromatin compaction and transcriptional silencing. Fig. 2. Histone structure modifications mediated by HAT and HDAC. The figure shows lysine at histone N-terminal tails have lysines suffering acetylation (HAT action) and deacetylation (HDAC action). In cellular pH, of approximately 7.4, the amine group is positively charged. When catalyzed by HAT, an acetyl group is added to its structure and the positive charge is neutralized. On the other hand, when HDAC acts an opposite mechanism occurs. Fig. 3. Hypothesis illustration of corepressor complexes involving HDAC. (A) Mad-Max require Sin3AHDAC to promote transcriptional repression. (B) Methylated CpG sites attract methyl-binding proteins such as MeCP2 that, in turn, attract Sin3A and HDAC for silencing transcriptional machinery.
Figr-1
Figr-2
Figr-3
Table 1. Summary of HDACs expression in human malignances. Protein
Tissue
Sample
Oral
49 patients
Lip
30 patients 91 patients
Lynphoma
Breast
Method
IHC
Reference
↑
[25]
↑
[5]
↓
[14]
↑
[1]
104 patients
IHC/WB
↑
[15]
200 patients
IHC/RT-PCR
Heterogeneous
[13]
20 patients
IHC/WB
↑
[18]
58 patients
IHC
↓
[24]
238/300 patients
IHC
Heterogeneous
[17],[22]
8 patients/2 cell lines
IF/WB
283 TMA
↑
[26]
Heterogeneous
[16]
↑
[28]
qRT-PCR/WB
↑
[7]
25 patients
IB/RT-PCR
↑
[4]
20 patients
IHC/WB
↑
[18]
102 patients
IHC/RT-PCR
↑
[21]
20 patients
IHC/WB
↑
[18]
28 patients
IHC/RT-PCR
↑
[19]
14 patients
IHC/FC/IF/WB
↑
[8]
20 patients
IHC/WB
↑
[18]
192 patients
IHC
↑
[27]
64 patients
IHC
↑
[10]
20 patients
IHC/WB
↑
[18]
20 patients
IHC/WB
↑
[18]
115 patients
IHC
↑
[9]
17 patients
IHC
↓
[12]
41 patients
IHC
↑
[2]
20 patients
IHC/WB
↑
[18]
57 patients
IHC
↑
[11]
20 patients
IHC/WB
↑
[18]
74 patients
IHC
↑
[6]
Esophageal
20 patients
IHC/WB
↑
[18]
Oral
93 patients
Leukemia
Gastric HDAC1 Lung
Prostate
Colorectal Ovary Endometrial Pancreatic Thyroid
94 patients 39 patients 24 cell lines 93 patients
49 patients Lip Lymphoma
Breast
HDAC2 Leukemia
Gastric Lung Prostate Pancreatic Colorectal, ovary, pancreatic, thyroid and esophageal
qRT-PCR
IHC
Lymphoma
[3]
↑
[25]
↑
[5]
IHC
↑
[1],[14]
104 patients
IHC/WB
↑
[15]
20 patients 8 patients 2 cell lines 58 patients
IHC/WB
↑
[18]
IF/WB
↑
[26]
30 patients
IHC
↓
[24]
Heterogeneous
238/300 patients 94 patients 39 patients/24 cell lines 93 patients
qRT-PCR
↑
qRT-PCR/WB
↑
[17], [22] [16] [28] [7]
71 patients
IHC
↑
[23]
20 patients
IHC/WB
↑
[18]
20 patients
IHC/WB
↑
[18]
20 patients
IHC/WB
↑
[18]
192 patients
IHC
↑
[27]
57 patients
IHC
↑
[11]
20 patients
IHC/WB
↑
[18]
8 patients/2 cell lines
IF/WB
↑
[26]
238 patients
↑ IHC
300 patients 20 patients
HDAC3
↑
283/91 patients
283 TMA
IHC/WB
Heterogeneous
[1] [17] [22]
↓
[18]
↑
104 patients 94 patients 39 patients/24 cell lines 20 patients
qRT-PCR
↑
IHC/WB
↑
[15] [16] [28] [18]
20 patients
IHC/WB
↑
[18]
192 patients
IHC
↑
[27]
Colorectal, ovary, pancreatic, thyroid and esophageal Lymphoma
20 patients
IHC/WB
↑
[18]
91 patients
IHC
↑
[14]
Breast
8 patients 2 cell lines
IF/WB
↓
[26]
Leukemia Lung Prostate
HDAC4
Expression
Leukemia
Endometrial HDAC5
Lymphoma Breast Leukemia
qRT-PCR
Heterogeneous
[16]
39 patients/24 cell lines 93 patients
94 patients
qRT-PCR/WB
↑ ↑
[28] [7]
41 patients 91 patients
IHC IHC
↑ ↑
[2] [14]
8 patients 2 cell lines 94 patients
IF/WB
↓
[26]
qRT-PCR
Heterogeneous
[16]
↑
[28]
39 patients/24 cell lines HDAC6
Pancreatic Oral
57 patients 9 patients
IHC IHC/IF/WB/qRT-PCR
↑ ↑
[11] [20]
Lymphoma
91 patients
IHC
↑
[14]
Breast
58 patients
IHC
↑
[24]
Heterogeneous
[22]
300 patients 8 patients/2 cell lines
IF/WB
↑
[26]
94 patients 39 patients/24 cell lines 8 patients 2 cell lines 94 patients
qRT-PCR
↑
IF/WB
↓
[16] [28] [26]
qRT-PCR
↑
[16]
41 patients 20 patients
IHC IHC/WB
↑ ↓
[2] [18]
8 patients 2 cell lines 94 patients 39 patients/24 cell lines 93 patients
IF/WB
↑
[26]
qRT-PCR
↑
RT-PCR/WB
↑
[16] [28] [7]
Gastric, lung, prostate, colorectal, ovary, pancreatic, thyroid and esophageal Breast
41 patients 20 patients
IHC IHC/WB
↑ ↑
[2] [18]
8 patients/2 cell lines
IF/WB/WB
↓
[26]
Leukemia
94 patients
qRT-PCR
Heterogeneous
[16]
↑
[28]
Leukemia HDAC7
Breast Leukemia Endometrial
HDAC8
Breast
Leukemia
Endometrial
HDAC9
39 patients/24 cell lines HDAC10
Breast
8 patients/2 cell lines
IF/WB
↑
[26]
Leukemia
94 patients
qRT-PCR
Heterogeneous
[16]
↑
[28]
39 patients/24 cell lines HDAC11
Breast
8 patients/2 cell lines
IF/WB
↓
[26]
Leukemia
94 patients
qRT-PCR
Heterogeneous
[16]
93 patients
qRT-PCR/WB
↑
[7]
57 patients
IHC
↑
[11]
Pancreatic
TSA: trichostatin A; VPA: valproic acid; SFN: sulforaphane; SAHA: suberoylanilide hydroxamic acid.
Gene
Pancreatic
HDAC1 / HDAC2 HDAC1 / HDAC2 HDAC1 / HDAC3 HDAC3 HDAC3 HDAC1
TSA
Phi Van et al.[13]
FoxO3
Nasopharyngeal Ovarian Gastric Gastric Hepatic, lymphoma, myelomonocytic tumor Breast
Inhibitor TSA / VPA / Butyrate Depsipeptide / TSA Valproate VPA SFN TSA TSA TSA TSA SAHA TSA TSA VPA TSA / Apicidin / SAHA TSA TSA
HDAC3
-
You et al. [17]
Survivin
Oral
-
TSA
Liu et al. [9]
TOP1
Colorectal
-
TSA
Meisenberg et al. [11]
p21
APC DAB2 GATA4 GATA6 MUC2 RI p53 E-cadherin DTWD1 PUMA 5-HTT
Cancer Cell lines Colon Lung, gastric, colon Lung Endometrial Colon Colon Ovarian Ovarian Ovarian Pancreatic Breast Lung
Target protein HDAC1 / HDAC2 HDAC1 / HDAC2 HDAC2 HDAC2 HDAC1 / HDAC2 HDAC1 / HDAC2 HDAC1 / HDAC2 HDAC1
Reference Huang et al.[6] Lin et al.[8] Zhao[18] Hrzenjak et al.[5] Zhu et al. [19] Myzak et al.[12] Caslini et al.[2] Caslini et al.[2] Caslini et al.[2] Yamada et al[16] Ammanamanchi e Brattain[1] Kim et al.[7] Von Burstin et al.[15] Tong et al.[14] Hayashi et al.[4] Ma et al.[10] Feng et al.[3]
TSA: trichostatin A; VPA: valproic acid; SFN: sulforaphane; SAHA: suberoylanilide hydroxamic acid.
Table 2. Examples of genes probably silenced by HDACs and inhibitors utilized for gene reexpression.
Table 3. Summary of the involvement of most studied HATs and their target genes in cancer. HAT Family
HAT
Gene E2F1, cyclin D1 and cyclin E1
GNAT
GCN5
P300
-
NKG2D
-
Cyclin E Rb
Rb P53 AR
Prostate cancer
c-Myc
T-cell leukemia/ Lymphoma
MYC
ZNF384
MYST HBO1 MOZ
Melanoma Lung and hematopoietic cancer Gastric cancer Lymphoblastic leukemia -
p53, caspase-3
Tip60
Lung cancer
KLF4
p53
P300/CBP
Favoring oncogenesis
NF-κB AR NF-κB VHL Nrf2 AML-1
Prostate cancer Liver cancer -
Favoring Tumor suppression
Reference
-
Chen et al. [3]
Esophageal Squamous Cell Carcinoma Breast/Colon/ Osteosarcoma/Melanoma Glioma, ovarian, breast, colorectal, lung and pancreatic cancer Colon and cervical cancer
Bandyopadhyay et al.[2] Iyer et al.[11]
-
Ogiwara et al.[15]
Hu et al. [9] Hu et al. [10] Gayther et al.[6]
-
Shi et al. [19]
-
Qian et al. [17]
Lung cancer Skin tumor
Leduc et al.[14] Hobbs et al.[8] Gaughan et al., 200296; Halkidou et al.[5, 7]
-
Awasthi [1]
Metastasis Leukemia Renal cancer Leukemia
Kim et al.[13] Sharma et al. [18] Contzler et al.[4] Zhou et al.[20] Ohta et al.[16] Katsumoto et al.[12]
Table 4. HDAC inhibitors approved by FDA and same researches involving these drugs.
HDACi
Vorinostat (SAHA, Zolina)
Romidepsin (Istodax, FK228, FR901228,
Class
Approvation year
Association
Tumor target
Reference
2006
-
Cutaneous T-cell Lymphoma
Mann et al. [20]
-
Hematologic malignancies
Kelly et al. [12]
Temozolomide and radiotherapy
Glioblastoma
Shi et al. [30]
-
Endometrial
Bokhman[3]; Lax[15]; Sarfstein et al.[28]
Hypoxia and radiotherapy
Lung
Ma et al.[17]
-
Gastrointestinal cancer
Saelen et al.[27]
AA98
Ovarian cancer
Ma et al.[18]
Cisplatin
Adenoid Cystic Carcinoma
Almeida et al. [1]
Cisplatin
Rhabdomyosarcoma
Jarzab et al.[9]
Bevacizumab
Clear-cell Renal Cell Carcinoma
Pili et al.[23]
Bortezomib
Multiple myeloma
Nanavati et al. [21]
2009
-
Cutaneous T-cell Lymphoma
Piekarz et al.[22]; Whittaker et al.[36]
2011
-
Peripheral T-cell Lymphoma
Coiffier et al.[5] Shustov et al. [31]
Bortezomib
Lung
Karthik et al.[11]
-
Metastatic Breast cancer
Robertson et al. [26]
Gemcitabine
Pancreatic, Breast, Lung, and ovarian
Jones et al. [10]
-
Thyroid
Amiri-Kordestani et al. [2]
-
Hepatocellular carcinoma
Sun et al. [32]
-
Peripheral T-cell Lymphoma
Poole [24]
-
Malignant pleural mesothelioma
Ramalingam et al. [25]
-
Ovarian tumors
Mackay et al. [19]
-
Thymic epithelial cancer
Giaccone et al. [8]
-
Mielodysplastyc syndrome
Cashen et al. [4]
Carboplatin and paclitaxel
Ovarian
Dizon, Blessing, et al.[6]; Dizon, Damstrup, et al.[7]
-
Acute myeloid leukemia
Kirschbaum et al. [13]
Cisplatin, doxorubicin, cyclophosphamide
Thymic epithelial tumors
Thomas et al. [33]
-
Liver
Ma et al. [16]
-
Acute promyelocytic leukemia
Savickiene et al. [29]
Hidroxamic acid In research
Cyclic peptide
depsipeptide) In research
2014
Belinostat (Beleodaq, PXD-101)
Hidroxamic acid In research
Doxorubicin
Soft Tissue Sarcomas
Vitfell-Rasmussen et al. [35]
Cisplatin
Squamous Cell Carcinoma
Kong et al. [14]
Cisplatin
Lung Cancer
To et al. [34]