Vous êtes sur la page 1sur 6

Molecular Pathways

Epigenetic Deregulation of DNA Repair and Its Potential for Therapy


Monika E. Hegi,1,2,3 Davide Sciuscio,1,2 Anastasia Murat,1,2 Marc Levivier,2 and Roger Stupp2

Abstract Epigenetic silencing of essential components of DNA repair pathways is a common


event in many tumor types, and comprise O6-methylguanine-DNA methyltransferase
(MGMT), human mut L homolog 1 (hMLH1), Werner syndrome gene (WRN), breast
cancer susceptibility gene 1 (BRCA1), and genes of the Fanconi anemia pathway. Most
interestingly, some of these alterations become the Achilles heel of the affected tumors
upon treatment with certain classes of anticancer agents. That is, patients whose tu-
mors carry such defects can be stratified for respective therapy rendering some classic
DNA damaging agents, such as alkylators or DNA crosslinking agents, into “targeted
therapies.” Here we review some of the affected repair pathways that, when inacti-
vated, sensitize the tumors to specific drugs and are thus exploitable for individualized
therapy. (Clin Cancer Res 2009;15(16):5026–31)

moter hypermethylation. Selection for aberrant DNA repair


Background
pathways in cancer development is in accordance with the con-
Epigenetic inactivation of genes by promoter methylation has cept that DNA damage signaling acts as a guardian against acti-
been recognized as an important mechanism by which tumor vated oncogenes and tumor progression (4). Thus, inactivation
suppressor genes are shut down during development of tumors. of such repair pathways represents important alterations in the
It represents one of the best studied mechanisms of aberrant development and malignant progression of these tumors. Genes
epigenetic modification in cancer. It is an integrated part of encoding proteins involved in DNA repair predominantly si-
the epigenetic regulatory system of gene expression involving lenced by promoter methylation in some tumor types comprise
changes in chromatin structure by post-translational modifica- MGMT (O6-methylguanine-DNA methyltransferase), hMLH1
tions of histones and nucleosome remodeling. Hypermethyla- (human mut L homolog 1), WRN (Werner syndrome gene),
tion of CpG islands in the promoter region leads to silencing and genes of the Fanconi anemia (FA) pathway such as FANCF,
either by direct inhibition of transcription factor binding, or FANCC, or FANCL (Fanconi complementation group) and in-
by attracting methylated-DNA binding proteins, recruiting oth- cluding BRCA1 (breast cancer susceptibility gene 1) (refs. 5–8).
er transcriptional repressors such as histone acetylases (HDACs) Most interestingly, some of the epigenetic alterations in these
and histone methyl transferases, resulting in transcriptionally repair pathways sensitize the affected tumors to treatment with
inactive chromatin. DNA methylation at the 5 position of cyto- certain classes of anticancer agents (Fig. 1).
sine is mediated by DNA methyltransferases (DNMTs; reviewed
in ref. 1). DNA Repair Pathways Affected by Epigenetic
Similar to other molecular aberrations, different tumor enti- Silencing in Tumors
ties exhibit their characteristic profiles of genes frequently affect-
ed by promoter hypermethylation (2). In accordance with the Epigenetically impaired repair by O6-methylguanine-DNA
Knudson 2-hit model of tumor suppression, methylation of methyltransferase. MGMT has become the most prominent
one allele is often accompanied by inactivation of the second of the epigenetically inactivated DNA repair genes, because
allele by deletion, mutation, or methylation (3). Genes encod- its promoter methylation has been associated with benefit
ing DNA repair proteins have been identified to become from alkylating agent therapy in malignant glioma (9), which
inactivated during development of several tumor types by me- was subsequently confirmed in clinical trials for newly diag-
chanisms such as deletion, mutation, and most commonly pro- nosed glioblastoma (10–13), and now serves as stratification
or inclusion criteria for patients entering clinical trials.
Evidence for prediction of benefit from multidrug regimens
Authors' Affiliations: 1Laboratory of Brain Tumor Biology and Genetics and comprising cyclophosphamide has been provided for B-cell
2
Department of Neurosurgery, University Hospital Lausanne (CHUV) and lymphoma (14).
University of Lausanne; and 3National Center of Competence in Research
MGMT rapidly reverses alkylation (including methylation) at
(NCCR) Molecular Oncology, ISREC SV-EPFL, Lausanne, Switzerland
Received 3/9/09; revised 3/16/09; accepted 3/25/09; published OnlineFirst the O6 position of guanine by transferring the alkyl-group to
8/11/09. the active site of the enzyme, constituted by a cysteine (15).
Requests for reprints: Monika E. Hegi, Laboratory of Brain Tumor Biology The DNA repair protein gets irreversibly inactivated in this
and Genetics, Department of Neurosurgery, Centre Hospitalier Universitaire suicide reaction, in fact this process is saturable, in that an ex-
Vaudois (CHUV BH19-110), 46 rue du Bugnon, Lausanne 1011, Switzerland.
Phone: 41-21-314-2582; Fax: 41-21-314-2587; E-mail: Monika.Hegi@chuv.ch.
cess of O6-methylguanine in the DNA can deplete MGMT in
F 2009 American Association for Cancer Research. the cells. Unlike other repair systems, MGMT acts alone to re-
doi:10.1158/1078-0432.CCR-08-1169 store guanine, and without formation of single strand breaks.

Clin Cancer Res 2009;15(16) August 15, 2009 5026 www.aacrjournals.org


Epigenetic Deregulation of DNA Repair and Therapy

Although, O6-alkyl-guanine is not the primary site of alkylation with an increased risk for acute myeloid leukemia, squamous
by alkylating chemotherapy agents, it seems to be the most cy- cell carcinoma of the head and neck, or gynecologic cancer. Re-
totoxic. An inactivated MGMT gene allows accumulation of this cent studies have revealed that proteins affected in this disease
lesion in the DNA, which subsequent to incorrect pairing with by mutation constitute a novel DNA-damage response network
thymidine triggers mismatch repair (MMR) thereby inducing that also involves the breast cancer proteins BRCA1 and BRCA2
DNA damage signaling and eventually cell death (16, 17). In (also known as FANCD1). Germline mutations in BRCA1 or 2
accordance with this mechanism MMR-deficient cells are highly are associated with a breast and ovarian cancer syndrome (re-
resistant to alkylating agents, even in the absence of MGMT. viewed in ref. 23).
Thus, MGMT rapidly repairs the most cytotoxic lesion of alky- In sporadic tumors the BRCA-Fanconi pathway is rarely inac-
lating agents, O6-alkyl guanine, thus reverting the therapeutic tivated by mutations, but frequently silenced by aberrant pro-
effect of alkylating drugs. Consequently, epigenetic inactiva- moter methylation of BRCA1 or FANCF and occasionally
tion of the MGMT gene by promoter methylation renders FANCC or FANCL. BRCA1 methylation has been identified in
MMR-proficient cells more sensitive to alkylating agents. In breast (11%–14%) and ovarian cancer (5%–31%), and infre-
accordance, mice overexpressing MGMT are more resistant to quently in lung and cervical carcinomas. FANCF promoter
carcinogenesis induced by alkylating agents, whereas respective methylation has been detected in ovarian (21%), breast
knock-out mice are more sensitive (18). (17%), head and neck cancers (15%), non-small cell lung can-
Tumors most commonly affected by epigenetic inactivation cer (14%), and cervical carcinomas (30%) (reviewed in ref. 8),
of MGMT comprise glioblastoma (45%), other glioma subtypes whereas FANCC and FANCL are infrequently methylated in leu-
(WHO grade I to III; 20 to <90%, subtype specific), colon kemia (24). In contrast to BRCA1 and FANCF, there is no evi-
(38%), lung (27%), and head and neck cancer (28%), and lym- dence for epigenetic silencing of BRCA2 in breast cancer (25),
phoma (25%) (refs. 5, 11, 14, 19). and it is only rarely present in ovarian cancers (26). No system-
The clinical relevance of epigenetic silencing of the MGMT atic studies are available linking epigenetic inactivation of the
promoter for benefit from alkylating agent therapy was shown BRCA-FA pathway with specific subtypes of breast cancer (27).
in a randomized trial for newly diagnosed glioblastoma. Pa- An intact BRCA-FA pathway is required to repair DNA cross-
tients whose tumors contained a methylated MGMT promoter links, stalled DNA replication forks, and double strand breaks.
had a clear survival benefit from the addition of the alkylating Eight of the BRCA-FA pathway proteins are subunits of an E3
agent temozolomide to standard radiotherapy with a median ligase (complex 1) that is required to mono-ubiquitinate
survival of 23.4 months [95% confidence interval (CI), 18.6– FANCD2, a critical step for the function of the BRCA-FA path-
32.8] as compared with 12.6 months (95% CI, 11.6–14.4.8) way. Mono-ubiquitinated FANCD2 interacts with BRCA2-
in patients with an unmethylated MGMT. In patients treated FANCD1 and other repair proteins to form DNA damage-
with initial radiotherapy only, no difference in progression-free inducible foci (complex 2). Consequently, loss of function of
survival could be shown, and a much smaller difference in over- any essential FA pathway component is expected to impair
all survival (11, 12). Similarly, and in concordance with the the pathway and predict sensitivity to DNA crosslinking agents
postulated predictive value of MGMT methylation, retrospective such as cisplatin, supported by experimental evidence (28). The
analyses of glioblastoma patients treated with alkylating agents cisplatin-sensitive human ovarian cancer cell line 2008 has a
showed a beneficial effect in presence of MGMT methylation, methylated FANCF promoter that upon prolonged exposure
whereas no association of MGMT status could be shown for pa- to cisplatin has been shown to become resistant, mediated by
tients treated with radiotherapy only (20, 21). restoration of FANCF expression associated with demethylation
New mechanistic in vivo evidence for the direct involvement of the FANCF gene. This in vitro observation suggests that selec-
of MGMT in glioblastoma response to alkylating agent therapy tion for demethylation of this gene, restoring of the FA path-
has been provided by the recent The Cancer Genome Atlas way, may account for acquired resistance to cisplatin
(TCGA) report on human glioblastoma (22). The mutation treatment in a clinical setting (29).
analysis of 601 genes in 91 matched tumor and/or normal sam- Tumors with inactivation of the BRCA-FA network may also
ples identified a hypermutator phenotype in the recurrent glio- respond to inhibitors of alternative DNA repair pathways.
blastoma of a subset of patients treated with alkylating agents, BRCA1 and BRCA2 are essential for the repair of double strand
which was confined to tumors with a methylated MGMT status breaks and stalled replication forks by homologous recombina-
in six of seven cases. Interestingly, the gene mutation pattern tion (HR). Cells deficient for BRCA1 and BRCA2 have been
was different in patients whose glioblastoma carried a methyl- shown to be exquisitely sensitive to poly(ADP-ribose) polymer-
ated MGMT promoter, compatible with the deficiency to repair ase (PARP) inhibitors (30, 31). Repair of damaged bases or re-
alkylated guanine residues. Moreover, in the six treated and gions of single-strand breaks through base excision repair
MGMT-methylated glioblastoma that were hypermutated, at (BER) requires PARP, a DNA-binding zinc finger protein that
least one of the MMR genes MLH1, MSH2, MSH6, or PMS2 catalyzes the transfer of ADP-ribose residues from NAD+ to itself
was mutated as compared with only one of 84 nonhypermu- and other proteins such as XRCC1, a key component thought to
tated, nontreated glioblastoma, which may suggest escape from initiate BER upon ADP-ribosylation (32). PARP-deficient, and
MGMT methylation-mediated sensitivity to the alkylating drug therefore BER-deficient mice, develop normally but have high
by selection for MMR deficiency. levels of sister chromatid exchange (a feature of HR),
Epigenetic inactivation of BRCA-Fanconi anemia pathway of suggesting that HR compensates for loss of PARP-dependent
DNA repair. The genomic instability syndrome FA is an auto- BER. Consequently PARP inhibitors are attractive drugs for
somal recessive disorder characterized by congenital abnormal- BRCA1- and 2-deficient tumors. In addition, PARP inhibitors
ities, bone marrow failure, cellular sensitivity to DNA-cross may also sensitize to alkylating agents such as temozolomide
linking agents, and a genetic cancer susceptibility syndrome as suggested in recent trials, because BER is an important

www.aacrjournals.org 5027 Clin Cancer Res 2009;15(16) August 15, 2009


Molecular Pathways

pathway repairing some of the lesions caused by these agents head and neck, breast, and colorectal cancer (6). However,
(reviewed in refs. 33, 34). hypermethylation of hMLH1 is often associated with other hy-
Epigenetic inactivation of WRN gene. Loss of function mu- permethylated genes, which complicates mechanistic interpre-
tations in WRN are the cause of an autosomal recessive disorder tation of associations with response to therapy in patients
characterized by premature aging, genomic instability, and pre- (39). Mechanistic investigations in vitro have shown that treat-
disposition to cancer. The WRN gene has been reported to be ment of hMLH1-methylated colon cancer cell lines with the
frequently silenced by DNA methylation in cancers of epithelial demethylating agent 5′aza-2′deoxycytidine (5-aza-dC) restores
and mesenchymal origin, most commonly in colorectal and hMLH1 expression and subsequently renders the cells MMR
non-small cell lung cancer (38%), gastric (25%) and prostate proficient (6). In accordance, 5-aza-dC treatment-mediated
cancer (20%), chondro-sarcomas (33%), and non-Hodgkin re-expression of hMLH1 overcomes in vitro resistance to 5-
lymphoma (23%; ref. 7). fluorouracil (5-FU) in colorectal cancer cell lines (40).
The WRN gene is ubiquitously expressed and belongs to the A new, but rare, mechanism of epigenetic inactivation of
RecQ helicase family of proteins with an intrinsic 3′–5′ exonu- MLH2 has been reported recently from families with Lynch syn-
clease activity. WRN is a major component of the DNA repair drome, exhibiting inheritable somatic methylation of the
and replication machinery, and has been shown to interact with MSH2 gene (41). This epigenetic inactivation by methylation
important DNA repair proteins such as DNA topoisomerase I, is mediated by deletion of the last exons of the immediate up-
BLM helicase, PARP-1, and RAD52. In line with the premature stream gene ACSTD1 in cis, and depends on the expression ac-
aging syndrome in absence of WRN, this helicase is involved in tivity of the deletion mutant. Expression of the truncated
telomere maintenance, and plays an important role in replica- ACSTD1, lacking a normal polyadenylation signal, results in
tive stress because of its function in repair, and remodeling transcriptional read-through into the MLH2 gene. In the affect-
and elongation of the replication fork (for review see ref. 35). ed patients, normal tissues positive for Ep-CAM (encoded by
Agrelo and colleagues reported that colorectal cancer cell ACSTD1), were associated with a methylated MLH2 gene in
lines with WRN promoter methylation are sensitive to the to- contrast to Ep-CAM negative tissues.
poisomerase inhibitor camptothecin and to mitomycin C. Most
interestingly, the authors showed that colorectal cancers exhi-
Clinical Translational Advances
biting WRN methylation indeed responded significantly better
to the topoisomerase I inhibitor irinotecan with a median over- Stratifying patients according to epigenetically impaired repair
all survival of 39.4 months versus 20.7 months (7). Sensitivity by MGMT and MGMT modulation. The clinical confirmation
to topoisomerase I inhibitors in WRN-negative cells has been that glioblastoma patients indeed benefit from alkylating agent
attributed to the conversion of treatment-induced single strand therapy dependent on the presumed nonfunctionality of DNA
breaks into double strand breaks at a high frequency in absence repair by MGMT, as determined by its promoter methylation
of WRN (36). Hypermethylation of WRN in colorectal tumors status, has led to a paradigm change in the treatment of these
could thus be a useful predictor of clinical response to topoi- patients. Whereas the association of MGMT methylation with
somerase I inhibitors. benefit from alkylating agent therapy is prospectively assessed,
Inactivation of mismatch repair through epigenetic inactivation attempts are taken to deplete the MGMT protein in the tumors
of hMLH1 is a factor in treatment resistance. The MMR gene of patients with an unmethylated MGMT gene using a dose dense
hMLH1 is an essential component of the DNA MMR pathway schedule of the alkylating agent (13),4 or treatment with non-
and is frequently mutated in hereditary nonpolyposis colon toxic inhibitors of the MGMT enzyme such as O6-benzylguanin,
cancer (HNPCC) also known as Lynch syndrome. Defects in or derivatives (42, 43). New trials select patients according to the
MMR-associated genes are common in many cancer types and re- MGMT methylation status in order to test new drugs in com-
sult in a mutator phenotype associated with microsatellite insta- bination with alkylating agent therapy expected to work best in
bility (37). The MMR system recognizes base–base mismatches patients with MGMT methylated tumors (NCT00689221);
and insertion or deletion loops (IDLs) in double-stranded (ds) patients with MGMT unmethylated tumors, thus proficient for
DNA, and degrades the region of the error-containing newly syn- repair by MGMT, are enrolled for studies testing new strategies
thesized strand, allowing the polymerase to correctly resynthe- based on molecular mechanisms other than DNA alkylation
size the second strand according to the template sequence. (NCT00509821). Another avenue is taken in the development
Activation of the MMR pathway may trigger DNA damage sig- of a new generation of alkylating drugs, yielding O6-guanine
naling, a process which induces cell cycle arrest and can lead to adducts that are not susceptible to repair by MGMT. However,
cell death in case of major DNA damages (for review see ref. 38). similar to strategies aiming at depleting MGMT in the tumor
In cancer, hMLH1 is the most commonly altered component of tissue using systemically delivered nontoxic MGMT inhibitors
this postreplicative DNA MMR system. It heterodimerizes with or high dose alkylating agent therapy, increased toxicity is
PMS2 to form MutL alpha, which gets recruited by MutS alpha expected, because the normal tissue is not protected by endo-
(MSH2–MSH6) or MutS beta (MSH2–MSH3), bound to a genous levels of MGMT.
dsDNA mismatch. Assembly of the MutL–MutS complex in pres- “BRCAness” and sensitivity to chemotherapy and PARP
ence of replication factor C (RFC) and proliferating cell nuclear inhibitors. Mechanism-based strategies targeting loss of func-
antigen (PCNA) is sufficient to activate endonuclease activity of tion of FA genes including BRCA2 comprise DNA cross-linking
PMS2. It introduces single-strand breaks near the mismatch and agents, whereas functional loss of BRCA1 also sensitizes to agents
thus generates new entry points for the exonuclease EXO1 to de- inducing double strand breaks. In accordance, the responsiveness
grade the strand containing the mismatch.
hMLH1 is the most prominent target of epigenetic silencing
in the MMR pathway in sporadic tumors, comprising ovarian, 4
http://clinicaltrials.gov, NCT00304031.

Clin Cancer Res 2009;15(16) August 15, 2009 5028 www.aacrjournals.org


Epigenetic Deregulation of DNA Repair and Therapy

Fig. 1. A, epigenetic silencing of DNA


repair pathways by promoter methylation
sensitizes tumors to specific therapy.
Genes encoding proteins involved in DNA
repair predominantly silenced by
promoter methylation comprise MGMT,
WRN, BRCA1, and genes of the FA
pathway such as FANCF, FANCC, or
FANCL (Fanconi complementation group).
B, in contrast, promoter methylation of
hMLH1 confers resistance to 5-FU in colon
cancer cells and to alkylating agents even
in the absence of MGMT. Combination
therapy with the DNA demethylating agent
5′-aza-2′deoxycytidine leads to
reexpression of hMLH1 and restores
sensitivity to 5-FU. Unmethylated CpGs,
open circles; methylated CpGs, black
circles.

of patients to chemotherapy depending on the BRCA1 promoter ation in many tumor types makes it an attractive biomarker for
methylation status is investigated in metastatic breast cancer with stratified use of topoisomerase I inhibitors that are widely used
a regimen combining cisplatin and bevacizumab (NCT00517361). as anticancer agents in many tumor types and potentially other
The sensitivity of “BRCAness” to inactivation of alternative repair DNA damaging drugs. The WRN promoter methylation status
pathways is evaluated with PARP inhibitors, as single agent or and benefit from therapy should be prospectively evaluated in
in combination with cisplatin, enrolling BRCA1-BRCA2- ongoing studies to establish a predictive or a prognostic value.
associated or hereditary breast and ovarian cancer patients Overcoming treatment resistance by restoring MMR using DNA
(NCT00664781, NCT00647062). However, methylated BRCA1/ demethylating agents. In contrast to epigenetic inactivation of
2 is not considered as inclusion criteria in these trials. the above-mentioned repair pathways, loss of MMR proficiency
Taking advantage of the epigenetically inactivated WRN through promoter methylation of hMLH1 leads to resistance to
gene. The reported high frequency of WRN promoter methyl- chemotherapy. Thus, reactivation of MMR by restoring hMLH1

www.aacrjournals.org 5029 Clin Cancer Res 2009;15(16) August 15, 2009


Molecular Pathways

expression using DNA demethylating agents such as 5-aza-dC is pathways that potentially render these tumors particularly sen-
an attractive approach. Azanucleoside drugs require incorpora- sitive to specific classes of therapeutic agents already in clinical
tion into the DNA to inhibit DNMT-catalyzed DNA methyla- use, or suggest novel avenues for drug development. Such strat-
tion. In accordance with this mechanism an ongoing trial for ified, personalized treatment strategies are supported by pre-
advanced ovarian, fallopian tube, and primary peritoneal can- clinical experiments. However, with the exception of MGMT
cer selects patients with methylated hMLH1 DNA in the plasma methylation in glioblastoma, little is known about epigenetic
for treatment with 5-aza-dC in combination with cisplatin inactivation of these repair pathways and their predictive value
(NCT00748527). Recently, 5-azacytidine and 5-aza-2′deoxycy- for benefit from therapies targeting their specific repair defects
tidine have been approved for myelodysplastic syndrome. In in larger cancer patient populations. Further, mechanistic corre-
addition to a DNA-demethylating activity 5-azacytidine also lations may be confounded by unknown associated alterations,
has a DNA-damaging effect inducing DNA repair signaling, e.g., a methylator phenotype, or conferring resistance such as an
which may account for part of its antitumor effect (44). MMR defect in MGMT-methylated cells treated with alkylating
An alternative strategy may be to specifically target cancers agents. In addition, it is not always clear if inactivation of both
in which the MMR repair pathway is perturbed. Compounds alleles of an affected gene is required to elicit the desired effect.
such as rhodium metalloinsertors bind to DNA base mis- These new treatment opportunities for individualized thera-
matches with high specificity and have been shown to inhibit py need to be translated into the clinical setting, which requires
cellular proliferation preferentially in MMR-deficient cells ver- robust biomarker tests for systematic molecular profiling of the
sus MMR-proficient cells (45). patients' tumors for subsequent patient selection and evalua-
tion in respective clinical trials.

Conclusion Disclosure of Potential Conflicts of Interest


As reviewed here, a number of tumor types exhibit clinically M.E. Hegi is a recipient of a commercial research grant from and a con-
interesting frequencies of epigenetically inactivated DNA repair sultant for Oncomethylome Sciences.

References
1. Jones PA, Baylin SB. The epigenomics of can- study: 5-year analysis of the EORTC-NCIC trial. 24. Hess CJ, Ameziane N, Schuurhuis GJ, et al.
cer. Cell 2007;128:683–92. Lancet Oncol 2009;10:459–66. Hypermethylation of the FANCC and FANCL pro-
2. Esteller M, Corn PG, Baylin SB, Herman JG. A 13. Glas M, Happold C, Rieger J, et al. Long-term moter regions in sporadic acute leukaemia. Cell
gene hypermethylation profile of human cancer. survival of patients with glioblastoma treated Oncol 2008;30:299–306.
Cancer Res 2001;61:3225–9. with radiotherapy and lomustine plus temozolo- 25. Collins N, Wooster R, Stratton MR. Absence
3. Herman JG, Baylin SB. Gene silencing in mide. J Clin Oncol 2009;27:1257–61. of methylation of CpG dinucleotides within the
cancer in association with promoter hyper‐ 14. Esteller M, Gaidano G, Goodman SN, et al. Hy- promoter of the breast cancer susceptibility
methylation. N Engl J Med 2003;349:2042–54. permethylation of the DNA repair gene O(6)- gene BRCA2 in normal tissues and in breast
4. Bartek J, Bartkova J, Lukas J. DNA damage methylguanine DNA methyltransferase and and ovarian cancers. Br J Cancer 1997;76:
signalling guards against activated oncogenes survival of patients with diffuse large B-cell 1150–6.
and tumour progression. Oncogene 2007;26: lymphoma. J Natl Cancer Inst 2002;94:26–32. 26. Hilton JL, Geisler JP, Rathe JA, Hattermann-Zogg
7773–9. 15. Pegg AE. Repair of O(6)-alkylguanine by alkyl- MA, DeYoung B, Buller RE. Inactivation of BRCA1
5. Esteller M, Hamilton SR, Burger PC, Baylin SB, transferases. Mutat Res 2000;462:83–100. and BRCA2 in ovarian cancer. J Natl Cancer Inst
Herman JG. Inactivation of the DNA repair gene 16. Ochs K, Kaina B. Apoptosis induced by 2002;94:1396–406.
O6-methylguanine-DNA methyltransferase D N A d a m a g e O6 - m e t h y l g u a n i n e i s B c l - 2 27. Schneider BP, Winer EP, Foulkes WD, et al.
by promoter hypermethylation is a common and caspase-9/3 regulated and Fas/caspase-8 in- Triple-negative breast cancer: risk factors to po-
event in primary human neoplasia. Cancer Res dependent. Cancer Res 2000;60:5815–24. tential targets. Clin Cancer Res 2008;14:8010–8.
1999;59:793–7. 17. Stojic L, Mojas N, Cejka P, et al. Mismatch 28. van der Heijden MS, Brody JR, Dezentje DA,
6. Herman JG, Umar A, Polyak K, et al. Incidence repair-dependent G2 checkpoint induced by et al. In vivo therapeutic responses contingent
and functional consequences of hMLH1 promot- low doses of SN1 type methylating agents re- on Fanconi anemia/BRCA2 status of the tumor.
er hypermethylation in colorectal carcinoma. quires the ATR kinase. Genes Dev 2004;18: Clin Cancer Res 2005;11:7508–15.
Proc Natl Acad Sci U S A 1998;95:6870–5. 1331–44. 29. Taniguchi T, Tischkowitz M, Ameziane N, et al.
7. Agrelo R, Cheng W-H, Setien F, et al. Epigenetic 18. Gerson SL. MGMT: its role in cancer aetiology Disruption of the Fanconi anemia-BRCA path-
inactivation of the premature aging Werner syn- and cancer therapeutics. Nat Rev Cancer 2004;4: way in cisplatin-sensitive ovarian tumors. Nat
drome gene in human cancer. Proc Natl Acad Sci 296–307. Med 2003;9:568–74.
U S A 2006;103:8822–7. 19. Mollemann M, Wolter M, Felsberg J, Collins 30. Farmer H, McCabe N, Lord CJ, et al. Target-
8. Turner N, Tutt A, Ashworth A. Hallmarks of VP, Reifenberger G. Frequent promoter hyper- ing the DNA repair defect in BRCA mutant
‘BRCAness’ in sporadic cancers. Nat Rev Can- methylation and low expression of the cells as a therapeutic strategy. Nature 2005;
cer 2004;4:814–9. MGMT gene in oligodendroglial tumors. Int J 434:917–21.
9. Esteller M, Garcia-Foncillas J, Andion E, et al. Cancer 2005;113:379–85. 31. Bryant HE, Schultz N, Thomas HD, et al. Spe-
Inactivation of the DNA-repair gene MGMT and 20. Criniere E, Kaloshi G, Laigle-Donadey F, et al. cific killing of BRCA2-deficient tumours with in-
the clinical response of gliomas to alkylating MGMT prognostic impact on glioblastoma is de- hibitors of poly(ADP-ribose) polymerase.
agents. N Engl J Med 2000;343:1350–4. pendent on therapeutic modalities. J Neuro- Nature 2005;434:913–7.
10. Hegi ME, Diserens AC, Godard S, et al. Clinical oncol 2007;83:173–9. 32. Hoeijmakers JH. Genome maintenance me-
trial substantiates the predictive value of O-6- 21. Zawlik I, Vaccarella S, Kita D, Mittelbronn M, chanisms for preventing cancer. Nature 2001;
methylguanine-DNA methyltransferase promoter Franceschi S, Ohgaki H. Promoter methylation 411:366–74.
methylation in glioblastoma patients treated with and polymorphisms of the MGMT gene 33. Lord CJ, Ashworth A. Targeted therapy for can-
temozolomide. Clin Cancer Res 2004;10:1871–4. in glioblastomas: a population-based study. cer using PARP inhibitors. Curr Opin Pharmacol
11. Hegi ME, Diserens AC, Gorlia T, et al. MGMT Neuroepidemiology 2009;32:21–9. 2008;8:363–9.
gene silencing and benefit from temozolomide 22. The Cancer Genome Atlas Research Net- 34. Plummer ER, Calvert H. Targeting poly(ADP-
in glioblastoma. N Engl J Med 2005;352:997–1003. work. Comprehensive genomic characteriza- ribose) polymerase: a two-armed strategy for
12. Stupp R, Hegi ME, Mason WP, et al. Effects of tion defines human glioblastoma genes and cancer therapy. Clin Cancer Res 2007;13:6252–6.
radiotherapy with concomitant and adjuvant core pathways. Nature 2008;455:1061–8. 35. Sidorova JM. Roles of the Werner syndrome
temozolomide versus radiotherapy alone on sur- 23. D'Andrea AD, Grompe M. The Fanconi anaemia/ RecQ helicase in DNA replication. DNA Repair
vival in glioblastoma in a randomised phase III BRCA pathway. Nat Rev Cancer 2003;3:23–34. (Amst) 2008;7:1776–86.

Clin Cancer Res 2009;15(16) August 15, 2009 5030 www.aacrjournals.org


Epigenetic Deregulation of DNA Repair and Therapy

36. Christmann M, Tomicic MT, Gestrich C, Roos with 5-fluorouracil based chemotherapy. Clin glioblastoma and clinical strategies to modulate
WP, Bohr VA, Kaina B. WRN protects against Cancer Res 2007;13:6093–8. MGMT activity. J Clin Oncol 2008;26:4189–99.
topo I but not topo II inhibitors by preventing 40. Strathdee G, MacKean MJ, Illand M, Brown R. 43. Quinn JA, Jiang SX, Reardon DA, et al. Phase
DNA break formation. DNA Repair (Amst) 2008; A role for methylation of the hMLH1 promoter II trial of temozolomide plus o6-benzylguanine in
7:1999–2009. in loss of hMLH1 expression and drug resistance adults with recurrent, temozolomide-resistant
37. Peltomaki P. Role of DNA mismatch repair de- in ovarian cancer. Oncogene 1999;18:2335–41. malignant glioma. J Clin Oncol 2009;27:1262–7.
fects in the pathogenesis of human cancer. J 41. Ligtenberg MJ, Kuiper RP, Chan TL, et al. Her- 44. Jiemjit A, Fandy TE, Carraway H, et al. p21
Clin Oncol 2003;21:1174–9. itable somatic methylation and inactivation of (WAF1/CIP1) induction by 5-azacytosine nucleo-
38. Jiricny J. The multifaceted mismatch-repair MSH2 in families with Lynch syndrome due to sides requires DNA damage. Oncogene 2008;27:
system. Nat Rev Mol Cell Biol 2006;7:335–46. deletion of the 3′ exons of TACSTD1. Nat Genet 3615–23.
39. Shen L, Catalano PJ, Benson AB III, O'Dwyer P, 2009;41:112–7. 45. Ernst RJ, Song H, Barton JK. DNA mismatch
Hamilton SR, Issa JP. Association between DNA 42. Hegi ME, Liu L, Herman JG, et al. Correlation of binding and antiproliferative activity of rhodium
methylation and shortened survival in O6-methylguanine methyltransferase (MGMT) metalloinsertors. J Am Chem Soc 2009;131:
patients with advanced colorectal cancer treated promoter methylation with clinical outcomes in 2359–66.

www.aacrjournals.org 5031 Clin Cancer Res 2009;15(16) August 15, 2009

Vous aimerez peut-être aussi