Skip to main content
Genes & Cancer logoLink to Genes & Cancer
. 2011 Jun;2(6):604–606. doi: 10.1177/1947601911423096

Cancer Epigenetics for the 21st Century

What’s Next?

Manel Esteller 1,2,3,
Editor: Manel Esteller
PMCID: PMC3174266  PMID: 21941616

Abstract

The discovery of global DNA hypomethylation events in human tumors in the early 1980s and the identification of CpG island promoter hypermethylation of tumor suppressor genes in cancer cells in the mid 1990s opened the door to the current excitement about the contribution of epigenetic disruption to human disease. The recent gigantic advances in technology make it possible to obtain complete DNA methylomes, histonomes, and non-coding RNA transcriptomes for many biological settings and their associated disorders. Furthermore, whole genome sequencing analyses yields an increasing number of mutated epigenetic genes in neoplasia. It is time to sit back, enjoy the show with a little help of friendly bioinformatic tools, and wonder about what will happen next.

Keywords: epigenetics, cancer, DNA methylation, histone, microRNA


There are many definitions for epigenetics, and that it can only mean that we are not sure about what the term means. One working definition might be “the inherited genome activity that does not depend on the naked DNA sequence.” Epigenetics also explains how the same genotype can produce different phenotypes as it occurs in monozygotic twins.1 There are many chemical modifications affecting DNA, RNA, and proteins that create the different epigenetic layers. Among the most studied epigenetic mechanisms, we can mention DNA methylation, histone modifications, and chromatin remodeling factors associated with nucleosome positioning. I also like to include non-coding RNAs as another level of epigenetic control, for their capacity to establish other epigenetic marks and control gene expression, but many “purists” will probably tear their hair out about this. But there is one thing for certain: the epigenetic setting is completely distorted in human cancer.2-4

Human tumors undergo a paradoxal DNA methylation change characterized by a global loss of DNA methylation, which takes place mostly at DNA-repetitive regions, and a gain of methylation at the promoter CpG islands of tumor suppressor genes such as hMLH1, BRCA1, VHL, Rb, p16Ink4a, p14ARF, and p15Ink4b. From a translational standpoint, the original discovery of the powerful predictor effect of MGMT hypermethylation in chemosensitivity5 and the initial observation that DNA hypermethylation events were easily detectable in biological fluids6 have generated hundreds of similar articles, engaged the biotechnology sector, and led to many clinical trials. We are on the verge of many biomedical uses in this area to complement classical techniques to monitor oncology patients. But there is more than the DNA methylation of the CpG islands located in the minimal 5′-promoters; good examples are downstream DNA methylation events for gene isoforms7 and neighboring CpG islands regions with lower CpG density such as CpG shores.8

Another twist to the story was the finding that not only protein-coding genes but also microRNAs with growth inhibitory function underwent DNA methylation-associated silencing in cancer cells.9,10 This is a rapidly expanding field, and the publications in this area have been increasing in the last 4 years. But there is more beyond and to come in the next period: microRNAs are just the tip of the iceberg of the non-coding RNA world, and many other ncRNAs might also undergo epigenetic aberrations. A good example is provided by the promoter CpG island hypermethylation-associated silencing of transcribed ultraconserved regions.11 It is worth keeping in mind that the miRNA machinery itself is starting to emerge as an alternative bona fide mutational target in tumors, such it has been shown for TARBP2, DICER1, and XPO5.12-14

However, one the greatest breakthroughs in this area has been the use of comprehensive DNA methylation microarrays and the recent establishment of the whole genome bisulfite sequencing. If early versions of DNA methylation microarrays has provided a recent peak within the contribution of DNA methylation to tissue biology and disease,15 we can just imagine how new platforms such as the 4,500,000 CpG site microarray16 might help our research. Of course, we will eventually be able to have complete DNA methylomes in many interesting samples as has been carried out in extremely interesting cases,17-19 but right now it is expensive. Among further novel seminal discoveries in the area of DNA methylation and cancer has been the realization that DNA methyltransferases themselves can also be genetically altered in malignancies, such as occur with DNMT3A20 and DNMT3B.21 From a more basic standpoint, the recent eruption of new chemical modifications in DNA that can regulate/mediate the classical 5-methylcytosine mark, such as 5-hydroxymethylcytosine22 and 5-carboxylcytosine,23 are extremely important issues that might be associated with prestigious international award recognition in 10 to 20 years.

DNA methylation is not an isolated epigenetic mark; it is linked with others such as histone modifications. The histone proteins are not only the good-for-packaging elements from most of the 20th century, but also critical regulators of gene expression. It is a complex scenario: many isoforms, different positions to modify, and many chemical marks (acetylation, methylation, phosphorylation, sumoylation, unbiquitination, . . .). It is clear, however, that human tumors contain a major disruption of the histone modification landscape.24,25 Many causes can explain this scenario, such as upstream mutations in oncogenes and tumor suppressor genes, but the histone modifiers are also target of mutations in cancer: from amplified histone methyltransferases,26 demethylases,27 or mutated deacetylases28 to the most recently described occurring in EZH229 and UTX30 and the chromatin-remodeling proteins ARID1A31 and PBRM1.32 The last ones were obtained thanks to the “big” genomics approaches. And this is not exhaustive list.

I would like to finish with a short reflection about epigenetic therapies. In our professional life as biomedical scientists, we are many times approached by patients that require solutions to their health problems. We are their hope. And epigenetic proteins and marks are good targets for the development of new anticancer drugs. The proof-of-principle provided by the approval of DNA demethylating agents and histone deacetylase inhibitors for the treatment of leukemia and lymphoma patients has been a critical turning point in the field that recognizes the task of many researchers. It has also been an eye-opener for large pharmaceutical companies, in that they now have new putative epigenetic drugs in their portfolios. In addition to a better selection of sensitive patients for each type of drug, we should think deeply about new targets and compounds such as inhibitors for histone methyltransferases,33,34 sirtuins,35 or histone kinases,36 or even enhancers of the production of tumor suppressor microRNAs.37 Only our imagination is the limit. Please regularly check for new developments in this area.

— Manel Esteller
Invited Editor

References

  • 1. Fraga MF, Ballestar E, Paz MF, et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci USA. 2005;102:10604-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128:683-92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Feinberg AP. Phenotypic plasticity and the epigenetics of human disease. Nature. 2007;447:433-40 [DOI] [PubMed] [Google Scholar]
  • 4. Rodríguez-Paredes M, Esteller M. Cancer epigenetics reaches mainstream oncology. Nat Med. 2011;17:330-9 [DOI] [PubMed] [Google Scholar]
  • 5. Esteller M, Garcia-Foncillas J, Andion E, et al. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 2000;343:1350-4 [DOI] [PubMed] [Google Scholar]
  • 6. Esteller M, Sanchez-Cespedes M, Rosell R, Sidransky D, Baylin SB, Herman JG. Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Res. 1999;59:67-70 [PubMed] [Google Scholar]
  • 7. Maunakea AK, Nagarajan RP, Bilenky M, et al. Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature. 2010;466:253-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Hansen KD, Timp W, Bravo HC, et al. Increased methylation variation in epigenetic domains across cancer types. Nat Genet. 2011;43:768-75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Saito Y, Liang G, Egger G, et al. Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell. 2006;9:435-43 [DOI] [PubMed] [Google Scholar]
  • 10. Lujambio A, Ropero S, Ballestar E, et al. Genetic unmasking of an epigenetically silenced microRNA in human cancer cells. Cancer Res. 2007;67:1424-9 [DOI] [PubMed] [Google Scholar]
  • 11. Lujambio A, Portela A, Liz J, et al. CpG island hypermethylation-associated silencing of non-coding RNAs transcribed from ultraconserved regions in human cancer. Oncogene. 2010;29:6390-401 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Melo SA, Ropero S, Moutinho C, et al. A TARBP2 mutation in human cancer impairs microRNA processing and DICER1 function. Nat Genet. 2009;41:365-70 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 13. Hill DA, Ivanovich J, Priest JR, et al. DICER1 mutations in familial pleuropulmonary blastoma. Science. 2009;325:965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Melo SA, Moutinho C, Ropero S, et al. A genetic defect in exportin-5 traps precursor microRNAs in the nucleus of cancer cells. Cancer Cell. 2010;18:303-15 [DOI] [PubMed] [Google Scholar]
  • 15. Fernandez AF, Assenov Y, Martin-Subero JI, et al. A DNA methylation fingerprint of 1628 human samples. Genome Res. 2011. Epub ahead of print [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Sandoval J, Heyn HA, Moran S, et al. Validation of a DNA methylation microarray for 450,000 CpG sites in the human genome. Epigenetics. 2011;6:692-702 [DOI] [PubMed] [Google Scholar]
  • 17. Lister R, Pelizzola M, Dowen RH, et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature. 2009; 462:315-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Li Y, Zhu J, Tian G, et al. The DNA methylome of human peripheral blood mononuclear cells. PLoS Biol. 2010;8:e1000533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Lister R, Pelizzola M, Kida YS, et al. Hotspots of aberrant epigenomic reprogramming in human induced pluripotent stem cells. Nature. 2011; 471:68-73 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Yan XJ, Xu J, Gu ZH, et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nat Genet. 2011;43:309-15 [DOI] [PubMed] [Google Scholar]
  • 21. Simó-Riudalbas L, Melo SA, Esteller M. DNMT3B gene amplification predicts resistance to DNA demethylating drugs. Genes Chromosomes Cancer. 2011;50:527-34 [DOI] [PubMed] [Google Scholar]
  • 22. Véron N, Peters AH. Epigenetics: Tet proteins in the limelight. Nature. 2011;473:293-4 [DOI] [PubMed] [Google Scholar]
  • 23. He YF, Li BZ, Li Z, et al. Tet-mediated formation of 5-carboxylcytosine and its excision by TDG in mammalian DNA. Science. 2011. Epub ahead of print [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Fraga MF, Ballestar E, Villar-Garea A, et al. Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat Genet. 2005;37:391-400 [DOI] [PubMed] [Google Scholar]
  • 25. Seligson DB, Horvath S, Shi T, et al. Global histone modification patterns predict risk of prostate cancer recurrence. Nature. 2005;435:1262-6 [DOI] [PubMed] [Google Scholar]
  • 26. Varambally S, Dhanasekaran SM, Zhou M, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419:624-9 [DOI] [PubMed] [Google Scholar]
  • 27. Cloos PA, Christensen J, Agger K, et al. The putative oncogene GASC1 demethylates tri- and dimethylated lysine 9 on histone H3. Nature. 2006;442:307-11 [DOI] [PubMed] [Google Scholar]
  • 28. Ropero S, Fraga MF, Ballestar E, et al. A truncating mutation of HDAC2 in human cancers confers resistance to histone deacetylase inhibition. Nat Genet. 2006;38:566-9 [DOI] [PubMed] [Google Scholar]
  • 29. Morin RD, Johnson NA, Severson TM, et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat Genet. 2010;42:181-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. van Haaften G, Dalgliesh GL, Davies H, et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet. 2009;41:521-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Jones S, Wang TL, Shih IeM, et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science. 2010;330:228-31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Varela I, Tarpey P, Raine K, et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. 2011;469:539-42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Tan J, Yang X, Zhuang L, et al. Pharmacologic disruption of Polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev. 2007;21:1050-63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Daigle SR, Olhava EJ, Therkelsen CA, et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell. 2011;20:53-65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Lara E, Mai A, Calvanese V, et al. Salermide, a Sirtuin inhibitor with a strong cancer-specific proapoptotic effect. Oncogene. 2009;28:781-91 [DOI] [PubMed] [Google Scholar]
  • 36. Huertas D, Soler M, Moreto J, et al. Antitumor activity of a small-molecule inhibitor of the histone kinase Haspin. Oncogene. 2011. Epub ahead of print [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Melo S, Villanueva A, Moutinho C, et al. Small molecule enoxacin is a cancer-specific growth inhibitor that acts by enhancing TAR RNA-binding protein 2-mediated microRNA processing. Proc Natl Acad Sci USA. 2011;108:4394-9 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Genes & Cancer are provided here courtesy of Impact Journals, LLC

RESOURCES