Cancer Biology and Signal Transduction The Role of Gene Body Cytosine Modi fi cations in MGMT Expression and Sensitivity to Temozolomide

The DNA repair protein O-methylguanine-DNA methyltransferase (MGMT) is known to play a role in sensitivity to temozolomide. Promoter hypermethylation of MGMT is commonly used to predict low expression levels of MGMT in gliomas, despite observed discordance between promoter methylation and protein levels. Here, we investigated the functional role of gene body cytosinemodification in regulating levels ofMGMT gene expression and sensitivity to temozolomide. In 91 human glioblastoma samples, we observed significant variation inMGMT expression levels in patientswith anunmethylated promoter,with higher levels of gene body cytosine modification correlating with higher gene expression levels. Furthermore, inducing hypomethylation across theMGMT gene body with decitabine corresponded with decreased levels ofMGMT gene expression in lymphoblastoid and glioblastoma cell lines, indicating an important functional role for gene body cytosine modifications in maintaining gene expression. We reasoned that the decrease in MGMT expression induced by decitabinemay render resistant glioblastoma cell linesmore sensitive to temozolomide. Consistent with this reasoning, we found that the MGMT-expressing glioblastoma cell lines exhibiting an unmethylated MGMT promoter that were pretreated with decitabine became significantly more sensitive to temozolomide. Overall, our results suggest a functional role for gene body cytosine modification in regulating gene expression ofMGMT and indicate that pretreating patients whose tumors have an unmethylatedMGMT promoter with decitabine before temozolomide treatment may increase their response to therapy.Mol Cancer Ther; 13(5); 1334–44. 2014 AACR. Introduction The O-methylguanine-DNA methyltransferase (MGMT) gene encodes for a DNA repair protein that repairs Oalkylguanine-DNA adducts (1). This type of DNA damage can occur after certain environmental exposures, such as tobacco-specific nitrosamines, and from DNA alkylating agents used for chemotherapy (2, 3). Temozolomide is an alkylating agent that was FDA approved to treat brain tumors in 2005, including anaplastic astrocytoma and glioblastoma multiforme, the most aggressive form of brain cancer (4). Only 5% to 10% of the methylation adducts on DNA resulting from temozolomide treatment are O-methylguanine, but if MGMT is not present to repair the damage, these adducts trigger DNA mismatch repair and are highly cytotoxic (5, 6). Upon repair, MGMT is irreversibly inactivated, thereby requiring new protein synthesis for additional repair (7). Consequently, epigenetic alterations that affect transcriptional activity of MGMT, such as promoter hypermethylation, can have a significant effect on the number ofO-guanine lesions that are repaired (8, 9). Patientswith gliomaswith an aberrantly hypermethylated MGMT promoter respond better to temozolomide treatment compared with patients with an unmethylatedMGMT promoter, because they lackMGMT protein expression (10, 11). Although approximately 40% to 70% of patients with gliomas have a methylatedMGMT promoter (9, 12–14), promotermethylationdoesnot always show a strong correlation with MGMT protein levels (13, 15). For example, one study investigating the correlation between MGMT promoter methylation and protein levels showed that 7 of 40 gliomas had an unmethylated promoter yet no detectable MGMT protein, and 16 of 40 gliomas had aberrant methylation at the promoter yet still had detectable MGMT protein (13). Therefore, additional mechanisms of transcriptional and translational regulation are likely affecting expression of MGMT. The role of gene body cytosine methylation and hydroxymethylation in regulating gene expression levels is not fully understood, but several studies have observed a positive correlation between gene body cytosine modifications and gene expression levels (16–18). ForMGMT in particular, gene body methylation was first shown to correlate with gene expression levels in 1992 (19). Two Authors' Affiliations: Committee on Cancer Biology, Department of Medicine, and Comprehensive Cancer Center, The University of Chicago; and Institute of Human Genetics, University of Illinois, Chicago, Illinois Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Authors: Lucy Godley, Room 7124, 900 E. 57th St., Chicago, IL 60637. Phone: 773-702-4140; Fax: 773-702-9268; E-mail: lgodley@medicine.bsd.uchicago.edu; and M. Eileen Dolan, edolan@medicine.bsd.uchicago.edu doi: 10.1158/1535-7163.MCT-13-0924 2014 American Association for Cancer Research. Molecular Cancer Therapeutics Mol Cancer Ther; 13(5) May 2014 1334 on June 21, 2017. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst February 25, 2014; DOI: 10.1158/1535-7163.MCT-13-0924

[1]  Erika L. Moen,et al.  Genetic and epigenetic variants contributing to clofarabine cytotoxicity. , 2013, Human molecular genetics.

[2]  Wei Zhang,et al.  Genome-Wide Variation of Cytosine Modifications Between European and African Populations and the Implications for Complex Traits , 2013, Genetics.

[3]  N. Heintz,et al.  MeCP2 binds to 5hmc enriched within active genes and accessible chromatin in the nervous system , 2012, Cell.

[4]  Tammy M. Havener,et al.  A genome-wide association analysis of temozolomide response using lymphoblastoid cell lines shows a clinically relevant association with MGMT , 2012, Pharmacogenetics and genomics.

[5]  M. Bernaudin,et al.  O-Methylguanine-DNA Methyltransferase (MGMT) Promoter Methylation and Low MGMT-Encoded Protein Expression as Prognostic Markers in Glioblastoma Patients Treated With Biodegradable Carmustine Wafer Implants After Initial Surgery Followed by Radiotherapy With Concomitant and Adjuvant Temozolomide , 2012 .

[6]  Peter A. Jones Functions of DNA methylation: islands, start sites, gene bodies and beyond , 2012, Nature Reviews Genetics.

[7]  J. Scandura,et al.  Phase 1 study of epigenetic priming with decitabine prior to standard induction chemotherapy for patients with AML. , 2011, Blood.

[8]  A. Jankowska,et al.  CpG methylation patterns and decitabine treatment response in acute myeloid leukemia cells and normal hematopoietic precursors , 2011, Leukemia.

[9]  F. Lyko,et al.  Azacytidine and Decitabine Induce Gene-Specific and Non-Random DNA Demethylation in Human Cancer Cell Lines , 2011, PloS one.

[10]  Joseph K. Pickrell,et al.  DNA methylation patterns associate with genetic and gene expression variation in HapMap cell lines , 2011, Genome Biology.

[11]  B. Christensen,et al.  DNA methylation, isocitrate dehydrogenase mutation, and survival in glioma. , 2011, Journal of the National Cancer Institute.

[12]  A. von Deimling,et al.  The Next Generation of Glioma Biomarkers: MGMT Methylation, BRAF Fusions and IDH1 Mutations , 2011, Brain pathology.

[13]  B. Zani,et al.  Biological rationale for the use of DNA methyltransferase inhibitors as new strategy for modulation of tumor response to chemotherapy and radiation , 2010, Molecular Cancer.

[14]  C. Sommer,et al.  MGMT activity, promoter methylation and immunohistochemistry of pretreatment and recurrent malignant gliomas: a comparative study on astrocytoma and glioblastoma , 2010, International journal of cancer.

[15]  R. Wilson,et al.  Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. , 2010, Cancer cell.

[16]  Chia-Lin Wei,et al.  Dynamic changes in the human methylome during differentiation. , 2010, Genome research.

[17]  Helen Brady,et al.  A Comparison of Azacitidine and Decitabine Activities in Acute Myeloid Leukemia Cell Lines , 2010, PloS one.

[18]  W. Curry,et al.  Racial, ethnic and socioeconomic disparities in the treatment of brain tumors , 2009, Journal of Neuro-Oncology.

[19]  Madeleine P. Ball,et al.  Corrigendum: Targeted and genome-scale strategies reveal gene-body methylation signatures in human cells , 2009, Nature Biotechnology.

[20]  J. Herman,et al.  Correlation of O6-methylguanine methyltransferase (MGMT) promoter methylation with clinical outcomes in glioblastoma and clinical strategies to modulate MGMT activity. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[21]  F. Lyko,et al.  Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine , 2008, International journal of cancer.

[22]  Tyson A. Clark,et al.  Evaluation of genetic variation contributing to differences in gene expression between populations. , 2008, American journal of human genetics.

[23]  E. Plimack,et al.  Combining epigenetic and cytotoxic therapy in the treatment of solid tumors. , 2007, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[24]  Andreas Schätzlein,et al.  Phase I and pharmacodynamic trial of the DNA methyltransferase inhibitor decitabine and carboplatin in solid tumors. , 2007, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[25]  Gabriele Schackert,et al.  Long-term survival with glioblastoma multiforme. , 2007, Brain : a journal of neurology.

[26]  J. Schramm,et al.  Optimization of quantitative MGMT promoter methylation analysis using pyrosequencing and combined bisulfite restriction analysis. , 2007, The Journal of molecular diagnostics : JMD.

[27]  L. Godley,et al.  The use of hypomethylating agents in the treatment of hematologic malignancies , 2007, Leukemia and Lymphoma.

[28]  M. Weller,et al.  Temozolomide: a milestone in the pharmacotherapy of brain tumors. , 2005, Future oncology.

[29]  R. Pazdur,et al.  Food and Drug Administration Drug Approval Summary: Temozolomide Plus Radiation Therapy for the Treatment of Newly Diagnosed Glioblastoma Multiforme , 2005, Clinical Cancer Research.

[30]  I. Ferrer,et al.  Prognostic Significance of O6-Methylguanine-DNA Methyltransferase Determined by Promoter Hypermethylation and Immunohistochemical Expression in Anaplastic Gliomas , 2005, Clinical Cancer Research.

[31]  R. Momparler Pharmacology of 5-Aza-2'-deoxycytidine (decitabine). , 2005, Seminars in hematology.

[32]  B. Bataille,et al.  Correlation of Clinical Features and Methylation Status of MGMT Gene Promoter in Glioblastomas , 2004, Journal of Neuro-Oncology.

[33]  Peter A. Jones,et al.  Demethylation of a hypermethylated P15/INK4B gene in patients with myelodysplastic syndrome by 5-Aza-2'-deoxycytidine (decitabine) treatment. , 2002, Blood.

[34]  M. Christmann,et al.  DNA repair in resistance to alkylating anticancer drugs. , 2002, International journal of clinical pharmacology and therapeutics.

[35]  S. Gerson Clinical relevance of MGMT in the treatment of cancer. , 2002, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[36]  S. Goodman,et al.  Hypermethylation of the DNA repair gene O(6)-methylguanine DNA methyltransferase and survival of patients with diffuse large B-cell lymphoma. , 2002, Journal of the National Cancer Institute.

[37]  Scar,et al.  Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. , 2000, The New England journal of medicine.

[38]  S. Hecht,et al.  DNA adduct formation from tobacco-specific N-nitrosamines. , 1999, Mutation research.

[39]  J. Herman,et al.  Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. , 1999, Cancer research.

[40]  S. Clark,et al.  High sensitivity mapping of methylated cytosines. , 1994, Nucleic acids research.

[41]  J. Costello,et al.  Graded methylation in the promoter and body of the O6-methylguanine DNA methyltransferase (MGMT) gene correlates with MGMT expression in human glioma cells. , 1994, The Journal of biological chemistry.

[42]  B. Kaina,et al.  Genomic differences between O6-methylguanine-DNA methyltransferase proficient (Mex+) and deficient (Mex-) cell lines: possible role of genetic and epigenetic changes in conversion of Mex+ into Mex-. , 1992, Biochemical and biophysical research communications.

[43]  A. Pegg,et al.  Mammalian O6-alkylguanine-DNA alkyltransferase: regulation and importance in response to alkylating carcinogenic and therapeutic agents. , 1990, Cancer research.

[44]  J. Uhm IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype , 2012 .

[45]  小峰 千明 Promoter hypermethylation of the DNA repair gene O6-methylguanine-DNA methyltransferase is an independent predictor of shortened progression free survival in patients with low-grade diffuse astrocytomas , 2004 .

[46]  M. Dolan,et al.  Structure, function, and inhibition of O6-alkylguanine-DNA alkyltransferase. , 1995, Progress in nucleic acid research and molecular biology.