We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Epigenetic-based companion diagnostics

    Leander Van Neste

    MDxHealth PharmacoDx, Technologiepark 4, 9052 Ghent, Belgium

    ,
    Gregory R Jones

    MDxHealth Inc., 302 E. Pettigrew Street, Suite 240, Durham, NC 27701, USA

    ,
    Wim Van Criekinge

    MDxHealth PharmacoDx, Technologiepark 4, 9052 Ghent, Belgium

    &
    James S Clark

    † Author for correspondence

    MDxHealth SA, Tour 5 GIGA niveau +3, Avenue de l’Hôpital 11, 4000 Liège, Belgium.

    Published Online:https://doi.org/10.2217/pme.11.70

    In current medical practice, when a patient is diagnosed with cancer the treating physician generally follows a standard protocol, assigning the treatment that gives a favorable response in the largest proportion of patients. However, in many individual instances this approach may not be the most effective solution and, typically, treatment is only initiated or altered once the cancer has actually started progressing. During this process, patients will lose treatment time waiting to start chemotherapy or will endure severe side effects associated with toxic chemotherapeutic treatments. While some patients are undertreated because current diagnostic methods cannot provide accurate enough information regarding the aggressiveness or drug response of their disease, others with nonaggressive forms of cancer are overtreated and unnecessarily undergo the side effects associated with chemotherapeutic treatment. Epigenetic markers have been widely investigated and are considered key regulators of cellular transcription. Histone modifications and DNA methylation have been demonstrated to play key roles in maintaining stem-cell-like states, cellular differentiation and cancer. In particular, DNA methylation is a frequent, abundant and stable cancer mark, with an inherent role in oncogenesis and tumor progression. In this article, the potential of DNA methylation as a companion diagnostic is assessed, illustrated by exploring some development paths. Epigenetic silencing of MGMT is a key example of how biomarker development, biological pathways and clinical utility come together, serving as a hallmark of epigenetic companion diagnostics.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    Bibliography

    • Spear BB, Heath-Chiozzi M, Huff J et al. Clinical application of pharmacogenetics. Trends Mol. Med.7,201–204 (2001).
    • Gomez A, Ingelman-Sundberg M. Pharmacoepigenetics: its role in interindividual differences in drug response. Clin. Pharmacol. Ther.85,426–430 (2009).
    • Roses AD. Pharmacogenetics and drug development: the path to safer and more effective drugs. Nat. Rev. Genet.5,645–655 (2004).
    • Tremblay KD, Saam JR, Ingram RS et al. A paternal-specific methylation imprint marks the alleles of the mouse H19 gene. Nat. Genet.9,407–413 (1995).
    • Okano M, Bell DW, Haber DA et al. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell99,247–257 (1999).
    • Wolf SF, Jolly DJ, Lunnen KD et al. Methylation of the hypoxanthine phosphoribosyltransferase locus on the human X chromosome: implications for X-chromosome inactivation. Proc. Natl Acad. Sci. USA81,2806–2810 (1984).
    • Gardiner-Garden M, Frommer M. CpG islands in vertebrate genomes. J. Mol. Biol.196,261–282 (1987).
    • Raisner RM, Hartley PD, Meneghini MD et al. Histone variant H2A.Z marks the 5´ ends of both active and inactive genes in euchromatin. Cell123,233–248 (2005).
    • Witcher M, Emerson BM. Epigenetic silencing of the p16(INK4a) tumor suppressor is associated with loss of CTCF binding and a chromatin boundary. Mol. Cell34,271–284 (2009).
    • 10  Jones PL, Veenstra GJ, Wade PA et al. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat. Genet.19,187–191 (1998).
    • 11  Rountree MR, Bachman KE, Baylin SB. DNMT1 binds HDAC2 and a new corepressor, DMAP1, to form a complex at replication foci. Nat. Genet.25,269–277 (2000).▪▪ Identification of the biological processes that lie at the basis of epigenetic silencing.
    • 12  Mariadason JM. HDACs and HDAC inhibitors in colon cancer. Epigenetics3,28–37 (2008).
    • 13  Yang XJ, Seto E. The Rpd3/Hda1 family of lysine deacetylases: from bacteria and yeast to mice and men. Nat. Rev. Mol Cell Biol.9,206–218 (2008).
    • 14  Ahringer J. NuRD and SIN3 histone deacetylase complexes in development. Trends Genet.16,351–356 (2000).
    • 15  McGarvey KM, Van Neste L, Cope L et al. Defining a chromatin pattern that characterizes DNA-hypemethylated genes in colon cancer cells. Cancer Res.68,5753–5759 (2008).▪ Matching key histone modifications to DNA hypermethylation patterns.
    • 16  Barski A, Cuddapah S, Cui K et al. High-resolution profiling of histone methylations in the human genome. Cell129,823–837 (2007).
    • 17  Cameron EE, Bachman KE, Myohanen S et al. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat. Genet.21,103–107 (1999).
    • 18  Zhang Y, Reinberg D. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev.15,2343–2360 (2001).
    • 19  Matarazzo MR, De Bonis ML, Strazzullo M et al. Multiple binding of methyl-CpG and polycomb proteins in long-term gene silencing events. J. Cell Physiol.210,711–719 (2007).
    • 20  Margueron R, Trojer P, Reinberg D. The key to development: interpreting the histone code? Curr. Opin. Genet. Dev.15,163–176 (2005).▪▪ Basic concepts of the histone code and epigenetic silencing.
    • 21  Zhang Y, Ng HH, Erdjument-Bromage H et al. Analysis of the NuRD subunits reveals a histone deacetylase core complex and a connection with DNA methylation. Genes Dev.13,1924–1935 (1999).
    • 22  Diaz NM. Laboratory testing for HER2/neu in breast carcinoma: an evolving strategy to predict response to targeted therapy. Cancer Control8,415–418 (2001).
    • 23  Morris CM. Chronic myeloid leukemia: cytogenetic methods and applications for diagnosis and treatment. Methods Mol. Biol.730,33–61 (2011).
    • 24  Bekaii-Saab T. KRAS testing in metastatic colorectal cancer: implications on the use of biologic agents. Clin. Colorectal Cancer8,135–140 (2009).
    • 25  Yi JM, Dhir M, Van Neste L et al. Genomic and epigenomic integration identifies a prognostic signature in colon cancer. Clin. Cancer Res.17,1535–1545 (2011).
    • 26  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.343,1350–1354 (2000).▪ The use of MGMT as a predictive biomarker for therapeutic response.
    • 27  Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. N. Engl. J. Med.349,2042–2054 (2003).▪▪ Review paper summarizing the basic concepts of epigenetic silencing and DNA methylation.
    • 28  Veigl ML, Kasturi L, Olechnowicz J et al. Biallelic inactivation of hMLH1 by epigenetic gene silencing, a novel mechanism causing human MSI cancers. Proc. Natl Acad. Sci. USA95,8698–8702 (1998).
    • 29  Palmisano WA, Divine KK, Saccomanno G et al. Predicting lung cancer by detecting aberrant promoter methylation in sputum. Cancer Res.60,5954–5958 (2000).
    • 30  Schuebel KE, Chen W, Cope L et al. Comparing the DNA hypermethylome with gene mutations in human colorectal cancer. PLoS Genet.3,1709–1723 (2007).
    • 31  Sharma SV, Lee DY, Li B, Quinlan MP et al. Chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell141,69–80 (2010).
    • 32  Tsai HC, Baylin SB. Cancer epigenetics: linking basic biology to clinical medicine. Cell Res.21,502–517 (2011).
    • 33  Flotho C, Claus R, Batz C et al. The DNA methyltransferase inhibitors azacitidine, decitabine and zebularine exert differential effect on cancer gene expression in acute myeloid leukemia cells. Leukemia23,1019–1028 (2009).
    • 34  Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene29,4741–4751 (2010).
    • 35  Hegi ME, Diserens A, Gorlia T et al.MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med.352,997–1003 (2005).
    • 36  Gerson SL. MGMT: its role in cancer aetiology and cancer therapeutics. Nat. Rev. Cancer.4,296–307 (2004)
    • 37  Nakagawachi T, Soejima H, Urano T. Silencing effect of CpG island hypermethylation and histone modifications on O6-methylguanine-DNA methyltransferase (MGMT) gene expression in human cancer. Oncogene22,8835–8844 (2003).
    • 38  Bhakat KK, Mitra S. CpG methylation-dependent repression of the human O6-methylguanine-DNA methyltransferase gene linked to chromatin structure alteration. Carcinogenesis24,1337–1345 (2003).
    • 39  Brock MV, Hooker CM, Ota-Machida E et al. DNA methylation markers and early recurrence in stage I lung cancer. N. Engl. J. Med.358,1118–1128 (2008).
    • 40  Belinsky SA, Liechty KC, Gentry FD et al. Promoter hypermethylation of multiple genes in sputum precedes lung cancer incidence in a high-risk cohort. Cancer Res.66,3338–3344 (2006).
    • 41  Belinsky SA, Klinge DM, Dekker JD et al. Gene promoter methylation in plasma and sputum increases with lung cancer risk. Clin. Cancer Res.11,6505–6511 (2005).
    • 42  Glöckner SC, Dhir M, Yi JM et al. Methylation of TFPI2 in stool DNA: a potential novel biomarker for the detection of colorectal cancer. Cancer Res.69,4691–4699 (2009).
    • 43  Harris RA, Wang T, Coarfa C et al. Comparison of sequencing-based methods to profile DNA methylation and identification of monoallelic epigenetic modification. Nat. Biotechnol.28,1097–1105 (2010).▪▪ Comparison of current sequencing techniques that can be used for epigenetic profiling.
    • 44  Budiman MA, Smith SW, Ordway JM. DNA methylation in personalized medicine. Per. Med.8,35–43 (2010).
    • 45  Meissner A, Mikkelsen TS, Gu H et al. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature454,766–770 (2008).
    • 46  Meissner A, Gnirke A, Bell GW et al. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res.33,5868–5877 (2005).
    • 47  Sandoval J, Heyn HA, Moran S et al. Validation of a DNA methylation microarray for 450,000 CpG sites in the human genome. Epigenetics6,692–702 (2011).
    • 48  Grunau C, Clark SJ, Rosenthal A. Bisulfite genomic sequencing: systematic investigation of critical experimental parameters. Nucleic Acids Res.29,E65-5 (2001).
    • 49  Ehrich M, Zoll S, Sur S et al. A new method for accurate assessment of DNA quality after bisulfite treatment. Nucleic Acids Res.35,e29 (2007).
    • 50  Bibikova M, Lin Z, Zhou L et al. High-throughput DNA methylation profiling using universal bead arrays. Genome Res.16,383–393 (2006).
    • 51  Ball MP, Li JB, Gao Y et al. Targeted and genome-scale strategies reveal gene-body methylation signatures in human cells. Nat. Biotechnol.27,361–368 (2009).
    • 52  Jacinto FV, Ballestar E, Esteller M. Methyl-DNA immunoprecipitation (MeDIP): Hunting down the DNA methylome. Biotechniques44,35–43 (2008).
    • 53  Fraga MF, Ballestar E, Montoya G et al. The affinity of different MBD proteins for a specific methylated locus depends on their intrinsic binding properties. Nucleic Acids Res.31,1765–1774 (2003).
    • 54  Serre D, Lee BH, Ting AH. MBD-isolated genome sequencing provides a high-throughput and comprehensive survey of DNA methylation in the human genome. Nucleic Acids Res.38,391–399 (2010).▪ Affinity-based sequencing method for ‘genome-wide’ epigenome analyses.
    • 55  Lai AY, Wade PA. Cancer biology and NuRD: a multifaceted chromatin remodelling complex. Nat. Rev. Cancer11,588–596 (2011).
    • 56  Fandy TE, Herman JG, Kerns P et al. 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. Blood114,2764–2773 (2009).
    • 57  Jowaed A, Schmitt I, Kaut O et al. Methylation regulates α-synuclein expression and is decreased in Parkinson’s disease patients’ brains. J. Neurosci.30,6355–6359 (2010).
    • 58  Daniilidou M, Koutroumani M, Tsolaki M. Epigenetic mechanisms in Alzheimer’s disease. Curr. Med. Chem.18,1751–1756 (2011).
    • 59  Ballestar E. Epigenetic alterations in autoimmune rheumatic diseases. Nat. Rev. Rheumatol.7,263–271 (2011).
    • 60  Renaudineau Y, Youinou P. Epigenetics and autoimmunity, with special emphasis on methylation. Keio J. Med.60,10–16 (2011).
    • 101  The microeconomics of personalized medicine www.mckinseyquarterly.com/The_microeconomics_of_personalized_medicine_2527
    • 102  Critical path reports www.fda.gov/ScienceResearch/SpecialTopics/CriticalPathInitiative/CriticalPathOpportunitiesReports/ucm2006653
    • 103  Reflection paper on methodological issues associated with pharmacogenomic biomarkers in relation to clinical development and patient selection www.ema.europa.eu/ema/index.jsp?curl=pages/news_and_events/news/2011/07/news_detail_001297.jsp&murl=menus/news_and_events/news_and_events.jsp&mid=WC0b01ac058004d5c1