Review
Post Screen
Epigenetic opportunities and challenges in cancer

https://doi.org/10.1016/j.drudis.2009.10.010Get rights and content

Epigenetic covalent modifications of DNA and chromatin proteins strongly affect gene expression and cellular activity, and epigenetic misregulation occurs in several diseases, especially cancer. First-generation drugs targeting the relatively promiscuous DNA methylation and histone acetylation modifiers have had successes in the treatment of haematological cancers. Second-generation drug programmes are in the discovery phase, targeting epigenetic enzymes with more tightly defined modes of action. This review highlights some of the challenges in identifying the most appropriate new targets and the issues that need to be addressed to facilitate the successful entry of second-generation epigenetic drugs into the clinic.

Section snippets

Epigenetics and cancer

There is an extraordinary degree of complexity in chromatin modifications and signalling, yet there is substantial enthusiasm for the development and implementation of epigenetic therapies in several human disease areas, especially oncology. What has led to these high expectations? Broadly, it is a combination of three factors: axiom, experience and prediction.

Second-generation cancer epitherapeutics – the challenges

As shown in Fig. 3, DNMT and HDAC inhibitors are already licensed for certain indications, but programmes targeting the enzymes that perform the more subtle chromatin modifications are all in the discovery phase. This is an inevitable consequence of the pace at which epigenetic targets have been identified: DNMTs were being extracted from mammalian livers by the early 1970s [46], yet LSD1, the first enzyme shown to demethylate methylated histones, was only identified in 2004 [38]. The rapid

Concluding remarks

Epigenetic control of gene regulation is a rapidly developing field of substantial potential, and oncology is likely to be the therapeutic application in which the fastest progress is made. However, if knowledge, know-how and clinical expertise remain in isolated silos, as is currently the case, epigenetic therapies will continue to reach the market but in an ad hoc and inefficient manner, which will fail to meet fully the needs of industry or patients. A more co-ordinated approach, involving

Acknowledgements

The authors thank Neil Pegg and David Knowles for helpful discussions.

References (60)

  • J. Marango

    The MMSET protein is a histone methyltransferase with characteristics of a transcriptional corepressor

    Blood

    (2008)
  • X.Y. Zhang

    The putative cancer stem cell marker USP22 is a subunit of the human SAGA complex required for activated transcription and cell-cycle progression

    Mol. Cell

    (2008)
  • A. Scoumanne et al.

    The lysine-specific demethylase 1 is required for cell proliferation in both p53-dependent and -independent manners

    J. Biol. Chem.

    (2007)
  • L.R. Jensen

    Mutations in the JARID1C gene, which is involved in transcriptional regulation and chromatin remodeling, cause X-linked mental retardation

    Am. J. Hum. Genet.

    (2005)
  • S. Kubicek

    Reversal of H3K9me2 by a small-molecule inhibitor for the G9a histone methyltransferase

    Mol. Cell

    (2007)
  • S.L. Berger

    An operational definition of epigenetics

    Genes Dev.

    (2009)
  • K. Konishi et al.

    Targeting aberrant chromatin structure in colorectal carcinomas

    Cancer J.

    (2007)
  • S.D. Taverna

    How chromatin-binding modules interpret histone modifications: lessons from professional pocket pickers

    Nat. Struct. Mol. Biol.

    (2007)
  • D.P. Bednarik

    Inactivation of the HIV LTR by DNA CpG methylation: evidence for a role in latency

    EMBO J.

    (1990)
  • D.C. Kaslow et al.

    DNA methylation stabilizes X chromosome inactivation in eutherians but not in marsupials: evidence for multistep maintenance of mammalian X dosage compensation

    Proc. Natl. Acad. Sci. U. S. A.

    (1987)
  • T. Mohandas

    Reactivation of an inactive human X chromosome: evidence for X inactivation by DNA methylation

    Science

    (1981)
  • S.F. Wolf et al.

    Studies of X chromosome DNA methylation in normal human cells

    Nature

    (1982)
  • Q. Zhao

    PRMT5-mediated methylation of histone H4R3 recruits DNMT3A, coupling histone and DNA methylation in gene silencing

    Nat. Struct. Mol. Biol.

    (2009)
  • K. Marushige

    Activation of chromatin by acetylation of histone side chains

    Proc. Natl. Acad. Sci. U. S. A.

    (1976)
  • M. Lachner

    Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins

    Nature

    (2001)
  • T.Y. Roh

    The genomic landscape of histone modifications in human T cells

    Proc. Natl. Acad. Sci. U. S. A.

    (2006)
  • J.M. Gilmore et al.

    Deciphering the combinatorial histone code

    Nat. Methods

    (2007)
  • A. Imhof et al.

    Modifications of the histone N-terminal domains. Evidence for an “epigenetic code”?

    Mol. Biotechnol.

    (2001)
  • T. Jenuwein et al.

    Translating the histone code

    Science

    (2001)
  • B.M. Turner

    Histone acetylation and an epigenetic code

    Bioessays

    (2000)
  • Cited by (47)

    • A brief survey on the application of metal-catalyzed azide–alkyne cycloaddition reactions to the synthesis of ferrocenyl-x-1,2,3-triazolyl-R (x = none or a linker and R = organic entity) compounds with anticancer activity

      2023, Coordination Chemistry Reviews
      Citation Excerpt :

      The subtle balance between acetylation and deacetylation of the histone proteins has a significant influence on chromatin compactness and gene expression. Histone deacetylation, which is catalyzed by histone deacetylases (HDACs), results in chromatin condensation and transcription silencing [145,146]. If these processes occur in cancer-transformed cells, they confer to the cells the ability to evade apoptosis and loss of proliferation control.

    • A novel class I histone deacetylase inhibitor, I-7ab, induces apoptosis and arrests cell cycle progression in human colorectal cancer cells

      2015, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      It has been evident that dysregulation of epigenetic modification is involved in genesis, progression, heterogeneity and acquired drug resistance of cancers [44,45]. Currently, the major epigenetic therapies for cancer applied in clinical practice involve inhibition of DNA methyltransferase (DNMT) and HDACs [1]. Mammalian HDACs are grouped into four classes based on their structural homology to their yeast homologues.

    • Computer-aided Molecular Design of Compounds Targeting Histone Modifying Enzymes

      2015, Computational and Structural Biotechnology Journal
      Citation Excerpt :

      In this direction, different studies have highlighted how the histone alterations contribute to the onset and growth of a variety of cancers [7,23,24,27,32–41], among other pathologies. Consequently, enzymes operating PTMs on histones are constituting attractive therapeutic targets for the development of new therapies [13,31,42–44]. It should be noted that, while the resulting effects on chromatin collectively depend on the ensemble of histone PTMs, these are operated by precise variations of physicochemical properties that we recently reviewed [17].

    • Physicochemical modifications of histones and their impact on epigenomics

      2014, Drug Discovery Today
      Citation Excerpt :

      As a consequence of the increasing interest in this topic, many recent articles and reviews have reported and described new types of modification, specific histone residues that undergo PTMs and enzymes that operate these modifications [17–20]. In addition, many other studies have elucidated the impact that PTMs have on chromatin and on the recruitment of chromatin regulators, as well as their clinicopathological relevance in human disease [8,10,18,21–29]. A further level of knowledge stems from the observation that the selectivity of histone PTMs varies depending on the kind of modification: for example, acetylation is the most promiscuous histone modification, and is always associated with transcriptional activity; by contrast, methylation has a high degree of selectivity and is associated with repression or transcription, depending on the methylated residue and the number of attached methyl groups [30,31].

    • A cyclodextrin-capped histone deacetylase inhibitor

      2013, Bioorganic and Medicinal Chemistry Letters
    View all citing articles on Scopus
    View full text