You are on page 1of 13

102 Current Molecular Pharmacology, 2012, 5, 102-114

Temozolomide: Mechanisms of Action, Repair and Resistance


Jihong Zhang1, Malcolm F.G. Stevens2 and Tracey D. Bradshaw*,2

1
Faculty of Life Science, Kunming University of Science and Technology, Kunming, Yunnan, 650093, China
2
Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, NG7 2RD, UK
Abstract: Glioblastoma multiforme is the most common aggressive adult primary tumour of the central nervous system.
Treatment includes surgery, radiotherapy and adjuvant temozolomide (TMZ) chemotherapy. TMZ is an alkylating agent
prodrug, delivering a methyl group to purine bases of DNA (O6-guanine; N7-guanine and N3-adenine). The primary
cytotoxic lesion, O6-methylguanine (O6-MeG) can be removed by methylguanine methyltransferase (MGMT; direct
repair) in tumours expressing this protein, or tolerated in mismatch repair-deficient (MMR-) tumours. Thus MGMT or
MMR deficiency confers resistance to TMZ. Inherent- and acquired resistance to TMZ present major obstacles to
successful treatment.
Strategies devised to thwart resistance and enhance response to TMZ, including inhibition of DNA repair mechanisms
which contribute to TMZ resistance, are under clinical evaluation. Depletion of MGMT prior to alkylating agent
chemotherapy prevents O6-MeG repair; thus, MGMT pseudosubstrates O6-benzylguanine and lomeguatrib are able to
sensitise tumours to TMZ. Disruption of base excision repair (BER) results in persistence of potentially lethal N7- and N3-
purine lesions contributing significantly to TMZ cytoxicity particularly when O6-MeG adducts are repaired or tolerated.
Several small molecule inhibitors of poly(ADP-ribose)polymerase-1 (PARP-1), a critical BER protein are yielding
promising results clinically, both in combination with TMZ and as single agent chemotherapy in patients whose tumours
possess homologous recombination DNA repair defects. Another validated, but as yet preclinical protein target,
mandatory to BER is abasic (AP) endonuclease-1 (APE-1); in preclinical tests, APE-1 inhibition potentiates TMZ activity.
An alternative strategy is synthesis of a molecule, evoking an irrepairable cytotoxic O6-G lesion. Preliminary efforts to
achieve this goal are described.
Keywords: Base excision repair, glioblastoma multiforme, methyltransferase, mismatch repair, O6-methylguanine
methylguanine, temozolomide.

INTRODUCTION administration [6], but labile above pH 7, with a plasma half-


life of 1.8 hours at pH 7.4 [7]. Thus, TMZ is rapidly
Malignant glioma is the most common adult primary absorbed intact, but then undergoes spontaneous breakdown
tumour of the central nervous system (CNS). Median to form monomethyl triazene 5-(3-methyltriazen-1-yl)-
survival from time of diagnosis is approximately 12 – 15 imidazole-4-carboxamide (MTIC). MTIC further reacts with
months [1-2]. Glioblastoma multiforme (GBM; grade IV water to liberate 5-aminoimidazole-4-carboxamide (AIC)
astrocytoma) is the most prevalent and aggressive adult and the highly reactive methyldiazonium cation (Fig. 1). The
primary brain tumour whose hallmark features include active species methyldiazonium cation preferentially
uncontrolled cellular proliferation, diffuse infiltration, methylates DNA at N7 positions of guanine in guanine rich
resistance to apoptosis, robust angiogenesis and rampant regions (N7-MeG; 70%), but also methylates N3 adenine
genomic instability [3]. The current standard of care for (N3-MeA; 9%) and O6 guanine residues (O6-MeG; 6%) [8-
newly diagnosed GBM patients includes surgery, 9].
radiotherapy and adjuvant temozolomide (TMZ) treatment,
There is a narrow pH window close to physiological pH
conferring a median survival time of 14.6 months compared
at which the whole process of TMZ prodrug activation to
with 12.2 months for patients receiving radiotherapy alone.
methyl group transfer can occur. Brain tumours possess a
Although TMZ (Temodar; Schering-Plough Corporation)
more alkaline pH compared with surrounding healthy tissue,
offers some hope to GBM patients, a best 5-year survival
a situation which favours prodrug activiation preferentially
rate of only 9.8% is achieved [4-5].
within tumour tissue [10].
TEMOZOLOMIDE PRODRUG ACTIVATION Thus TMZ is used to treat (but not exclusively) brain
tumours, imparting significant therapeutic benefit to GBM
TMZ, a small (194 Da) lipophilic molecule (Fig. 1), is an patients [4].
orally available monofunctional DNA alkylating agent of the
imidazotetrazine class. TMZ acts as a prodrug, stable at TEMOZOLOMIDE CYTOTOXICITY
acidic pH values, a property which permits oral
Temozolomide cytotoxicity is primarily mediated
through O6-MeG, a carcinogenic, mutagenic and toxic lesion
*Address correspondence to this author at the Centre for Biomolecular [11-14]. Direct repair of O6-MeG by the suicide enzyme
Sciences, School of Pharmacy, University of Nottingham, NG7 2RD, UK; methylguanine-DNA methyltransferase (MGMT) removes
Tel: +44(0)115 9513419; Fax: +44(0)115 9515102;
E-mail: tracey.bradshaw@nottingham.ac.uk the methyl adduct, restoring guanine (Fig. 2). Unrepaired

1874-4672/12 $58.00+.00 © 2012 Bentham Science Publishers


Temozolomide: Mechanisms of Action, Repair and Resistance Current Molecular Pharmacology, 2012, Vol. 5, No. 1 103

H2NOC
H2NOC H N
N N CH3
N H2O N
N
N N - CO2 N H+
CH3 NH

H2NOC
NH2
NuH
N + CH3N2 NuCH3 + N2 + H+
NH
(Nu = nucleophilic centre on DNA)

Fig. (1). Structure and activation route of prodrug temozolomide.

O6-MeG mispairs with thymine (not cytosine) during DNA DNA REPAIR MECHANISMS CONTRIBUTING TO
replication, alerting DNA mismatch repair (MMR) [15-16]. TEMOZOLOMIDE RESISTANCE
MMR exclusively recognises the mispaired thymine on the
daughter strand and excises it, yet O6-MeG persists in the Direct Repair
template strand. Therefore, futile cycles of thymine re-
insertion and excision result in persistent DNA strand MGMT (O6-Alkylguanine-DNA alkyltransferase; AGT)
breaks, causing replication fork collapse [17]. G2/M cell repairs O6-alkylguanine adducts in a single step,
cycle arrest is triggered, occurring in the second cell cycle independently of any other protein or cofactors (Fig. 2). It is
following treatment [18-20] via ATR/CHK1-dependent a small protein (22 kDa) present in both the cytoplasm and
signalling [21]; ultimately, apoptosis ensues [22] (Figs. 3 and nucleus. Upon DNA alkylation, a shift towards more nuclear
4). A good response to TMZ therefore requires functional localisation may facilitate the repair process [24]. MGMT is
MMR and low levels of MGMT. able to repair not only O6-MeG, but also guanine residues
with longer O6-alkyl adducts such as ethyl, chloroethyl,
The quantitatively more abundant N7-MeG and N3-MeA hydroxyethyl, n-propyl, n-butyl, and more bulky cyclic
lesions are rapidly repaired by DNA base excision repair lesions conferred by benzyl or pyridyloxobutyl groups, but
(BER; Figs. 4 and 5). N7-MeG appears not to be markedly with diminishing efficiency as adduct size increases [25-27].
cytotoxic: in contrast, N3-MeA lesions are lethal if not The O6-alkyl group is transferred from guanine to the active
intercepted [23]. site cysteine residue (Cys 145) of MGMT in a
Therefore, the most important DNA repair systems stoichiometric, auto-inactivating reaction, thereby repairing
impacting the mechanism of action and cytotoxicty of TMZ DNA and inactivating MGMT [28]. MGMT binds damaged
are MGMT (direct repair), MMR and BER (Figs. 4 and 5). substrate DNA in the minor groove, the target base is then

MGMT MGMT

Cysteine 145 Cysteine 145

CH3
H2NOC
N
N CH3
N O
N N O
CH3 6 6
N N
N HN
O
H H
TMZ N
H2N N H2N N N

d Ribose d Ribose

O6-methylguanine guanine
Fig. (2). Repair of O6-methylguanine adducts by O6-methylguanine-DNA methyltransferase.
104 Current Molecular Pharmacology, 2012, Vol. 5, No. 1 Zhang et al.

A B
600

500 MGMT

SNB19M

U373V

U373M
SNB19V
400
GI50 (mm)

300

200

100

0
S N B 19V S N B 19M U 373V U 373M

Cell Line

C Control 24 h 48 h 72 h 120 h

Fig. (3). A) Effect of temozolomide on glioblastoma multiforme cell growth. Cells were seeded at a density of 650 per well. After 24 h,
temozolomide was introduced. At the time of drug addition and following 7 days incubation, MTT assays were performed to determine cell
growth.
B) Expression of methylguaninemethyltransferase protein in SNB19M and U373M cells. Western blot assays were performed following
separation of protein from whole cell lysates.
C) Effect of temozolomide on SNB19V cell cycle.

MGMT+ G survival

O6-Me-G
MMR+ cytotoxicity
MGMT-
TMZ MMR-

N7-,N3-Me-Purine mutations tolerated

BER survival

Fig. (4). Key DNA repair mechanisms influencing cellular response to temozolomide.

flipped out of the helix and bound to MGMT, altering the alkylating agents such as TMZ. Tissue expression is
conformation of the DNA binding domain allowing variable, with high protein expression in liver and lower
alkylated MGMT to be detached from DNA and degraded expression in haematopoietic tissues and brain [30-31].
through the ubiquitin/proteasomal system [15, 29]. Tumour MGMT expression is immensely variable, highest
levels are found in breast, ovarian and lung tumours,
MGMT protects cells from carcinogens; however, it is
whereas lowest activity is observed in gliomas, pancreatic
also able to protect cancer cells from chemotherapeutic
Temozolomide: Mechanisms of Action, Repair and Resistance Current Molecular Pharmacology, 2012, Vol. 5, No. 1 105

TMZ

O6-MeG N7-MeG N3-MeA


DR MMR BER

MGMT MSH2-MSH6 Glycosylase


MSH2-MSH3
(Mutsα and β) APE-1

MLH1-PMS2 PARP
MLH1-PMS1
(MutlLα and β)
short patch long patch
Nucleases (EXO1) Polβ Pol
RFC, PCNA, XRCC1 PCNA
RPA, DNA Pol Ligase III FEN1
Ligase Ligase 1
Fig. (5). Summary of proteins involved in DNA repair pathways activated by temozolomide-induced DNA lesions.

carcinomas and malignant melanomas [32]. Hence, TMZ (resulting from gains or losses of short repeat units within
treatment of metastatic malignant melanoma has been under microsatellite sequences) generated during DNA synthesis.
clinical evaluation [33] and is now an approved indication in MutS (comprising MSH2 and MSH6) or MutS
certain territories. In gliomas, however, MGMT activity (comprising MSH2 and MSH3) complexes recognise and
varying 300-fold has been reported [34], and a strong bind to mismatch lesions. MutS binds base to base
positive correlation exists between MGMT activity and mismatches and insertion/deletion mismatch loops of one or
alkylating agent resistance [35-37] in vivo and in vitro, as two nucleotides. Muts has little affinity for base to base
exemplified in Fig. (3). MGMT transfected SNB19 and mismatches [50-51], but is involved in repair of loops of up
U373 GBM cells are > 13- and > 5-fold more resistant to to 16 nucleotides. In man, Muts is predominantly
TMZ challenge than their isogenic, vector control transfected implicated in DNA damage signalling. The MSH2/MSH6
partner cell lines, possessing negligible and low MGMT heterodimer undergoes an ATP-dependent conformational
activity respectively. The MGMT gene is located on change and recruits the MLH1/PMS2 heterodimer, which
chromosome 10q26 and frequent loss of chromosome 10 coordinates the interplay between the mismatch recognition
observed in 60% – 85% glioma cases is thought to be related complex and additional proteins including exonuclease 1,
to low protein expression [38]. Mutations of MGMT and helicases, proliferating cell nuclear antigen, single strand
protein phosphorylation, responsible for MGMT inactivation DNA binding protein, DNA polymerase  and , necessary
have also been detected in human tumours [39]. However, for removal and replacement of the mismatched DNA base.
loss of MGMT activity is most frequently a consequence of MMR plays a critical role in correction of replicative
MGMT promoter methylation [40-42]. Gene inactivation by mismatches that have escaped polymerase proofreading, and
promoter methylation is a common epigenetic phenomenon loss of MMR results in a dramatic increase in insertion/
in tumourigenesis [43]. Methylation, mediated by 5`- deletion mutations, particularly in repetitive sequence
methylcytosine methyltransferase, takes place on the microsatellite DNA. Indeed, microsatellite instability (MSI)
cytosine of CpG islands. Hypermethylation of CpG islands is a recognised surrogate biomarker for the loss of MMR
in the MGMT promoter region prevents transcription factor function [52].
binding, silencing the gene [41, 44]. MGMT methylation has
MMR is of clinical significance in several cancers
been detected in 45% - 70% high grade gliomas [45-46].
including endometrial, ovarian, gastric and particularly
Clinical evidence has revealed that patients with MGMT colorectal cancers. Indeed, MMR deficiency has been
promoter methylation respond better than those without
observed in 15% - 20% of sporadic colorectal tumours [53-
promoter methylation to radiotherapy treatment with either
54] as well as some breast, prostate, bladder, head and neck
BCNU or TMZ [47-49]. The correlation between MGMT
cancers [55]. Colorectal cancer studies have demonstrated
promoter methylation extent and clinical response to
hereditary and sporadic MMR gene mutations responsible
alkylating agents means that MGMT promoter methylation is
for MSI. In hereditary nonpolyposis colorectal cancer
a good predictive marker of response to alkylating agent (HNPCC), germ line mutations in MLH1 or MSH2 cause
chemotherapy.
microsatellite repeat replication errors to persist [56].
Somatic MMR gene mutations may be the result of
DNA Mismatch Repair epigenetic gene silencing via methylation of the MLH1
Mismatch repair (MMR; Fig. 5) is the recognition and promoter [57-58]. Aberrantly methylated hMLH1 promoter
correction of mispaired bases and insertion/deletion loops DNA has been reported in the sera of 9/19 patients with
106 Current Molecular Pharmacology, 2012, Vol. 5, No. 1 Zhang et al.

microsatellite unstable colon carcinoma [59]. MMR polyribosylated PARP from the DNA lesion allows access of
mutations allow microsatellite insertions/deletions which essential BER proteins. Thus, PARP facilitates efficient
cause inactivating frameshift mutations within tumour DNA repair and survival of cells subjected to mild genotoxic
suppressor coding regions, critical genes in cell cycle stress [67]. Inhibition of PARP increases the frequency of
regulation and cancer prevention. DNA strand breaks, accordingly PARP deficient cells are
hypersensitive to carcinogenic agents [68].
MMR status influences the response of cells to genotoxic
agents, indeed, methylating agent cytotoxicity induced by As noted, the majority of DNA lesions generated by
TMZ requires functional MMR. HCT 116 (MLH1 mutant) TMZ are N7-MeG and N3-MeA, comprising 80-85% and 8-
and DLD1 (MSH6 mutant) MMR deficient colon carcinoma 18% of total alkyl adducts respectively. These lesions,
cells are resistant to TMZ treatment (GI50 > 500 μM) [60]. rapidly and efficiently repaired by BER, become highly
MMR-deficient cells are reported to be up to 100-fold less cytotoxic when BER is disrupted [69]. PARP inhibition
sensitive to methylating agents compared with their MMR enhances the cytotoxicty of base lesions normally repaired
proficient counterparts [21, 61]. In such cells, O6-MeG- by BER, and indeed, enhances TMZ cytotoxicity in vitro and
thymine mispairs are not recognised, O6-MeG lesions are in vivo [70-72]. TMZ cytotoxicity is primarily manifest via
tolerated, cells continue cycling, surviving at the expense of O6-MeG adducts. However, when O6-MeG lesions are
extensive mutagenesis [61]. either repaired by MGMT or tolerated following MMR
disruption, N-Me purine adducts become significant, and
Base Excision Repair then inhibition of BER enhances TMZ therapeutic efficacy
[70]. Therefore, disruption of BER by PARP inhibition
Base excision repair (BER) is the major pathway
provides a means to overcome resistance that frequently
involved in removal and repair of non-bulky damaged
develops as a consequence of selection of MMR deficient
nucleotides, abasic sites and DNA single strand breaks
cells during therapy [73].
generated by reactive oxygen species, ionising radiation and
alkylating agents [62]. N7- and N3- purines, methylated by
TMZ are repaired by BER (Figs. 4 and 5). Damaged bases Acquired Drug Resistance
are recognised by lesion-specific glycosylases that hydro- Tumours initially sensitive to chemotherapy often
lytically cleave the N-glycosidic bond, generating an abasic develop resistance – acquired resistance. Acquired drug
(apurinic/apyrimidinic; AP) site. AP endonuclease (APE-1) resistance emerges, by Darwinian evolution, as a
then cleaves the phosphodiester backbone on the 5`side of consequence of selective pressure in the presence of
the AP site, leaving 3`-OH and 5`-deoxyribose phosphate chemotherapeutic agent. Acquired chemo-resistance may be
(dRp) termini at the DNA strand break. The terminal 5`dRp a consequence of drug-induced genetic and epigenetic
residue is removed by exonuclease or DNA-deoxyribopho- changes in neoplastic cells, inducing and selecting genes
sphodiesterase, leaving a nucleotide gap. Further repair can
which confer a survival advantage, or result from selection
proceed via two pathways: short patch involving
of pre-existing resistant cell clones [74]. In initially
replacement of one nucleotide; and long patch involving gap
heterogenous tumours, chemotherapy eliminates drug-
filling of 2 – 10 nucleotides. In short patch BER, the single
sensitive malignant cells allowing survival of drug-resistant
nucleotide gap is filled by DNA polymerase and the nick
cells and chemo-resistant cancer stem cells which may
sealed by the DNA ligase III/X-ray repair cross-
advance to seed more (acquired) resistant tumours. Indeed,
complementing 1 (XRCC1) heterodimer [63]. In long patch
within glioblastomas and medullbalstomas, populations of
BER, the DNA strand may become displaced, creating a
cells with stem cell-like properties, associated with tumour-
flap. Flap endonuclease-1 (FEN-1) cleaves the flap and DNA
initiating capacity and resistance to therapy have been
ends are sealed by DNA ligase I [64].
identified [2, 75-76]. Thus tumours demonstrating resistance
A protein key to successful DNA damage signaling and to multiple chemotherapeutic agents whose mechanisms of
BER is poly(ADP-ribose) polymerase-1 (PARP-1). action are distinct may emerge [77]. Such multidrug
Constitutively expressed, but activated in response to DNA, resistance is a major factor contributing to treatment failure.
damage, PARP-1 modifies nuclear proteins by poly(ADP- Mechanisms conferring resistance to chemotherapeutic
ribosyl)ation. PARP-1 enzyme (113 kDa), encoded by the agents include decreased cellular drug uptake, increased drug
ADP-ribosyl transferase (ADPRT) gene, possesses 3 efflux by membrane pumps actively expelling chemotherapy
domains. The DNA binding domain, possessing 2 zinc finger agents, intracellular drug inactivation, alteration of drug
structures and a nuclear localisation sequence at the N- target by mutation, inactivation or over-expression, enhanced
terminus, recognises DNA single and double strand nicks repair of drug-induced DNA damage or suppression of repair
(SSBs and DSBs). The automodification domain mediates resulting in tolerance to DNA lesions and alteration of
protein/protein interactions, and at the C-terminus is the apoptosis-related genes [78-80]. The consequence of such
active site catalytic domain. In response to DNA damage, measures is survival of malignant cells resistant to drug-
PARP-1 binds to DNA SSB (or DSB) and cleaves – induced apoptosis
nicotnamide adenine dinucleotide (NAD+) releasing
nicotinamide and ADP-ribose. PARP uses NAD+ to catalyse MECHANISMS OF GLIOMA CHEMORESISTANCE
auto-(and other protein) poly(ADP-ribosyl)ation. Long, AND ACQUIRED DRUG RESISTANCE TO TEMO-
branched ADP-ribose polymers attract recruitment of the ZOLOMIDE
BER protein complex comprising XRCC1, DNA polymerase
 and DNA ligase III, FEN-1 to progress repair (and serve as Although TMZ is first line chemotherapy for glioma
an energy source for ligation) [65-66]. Release of patients, inherent and acquired resistance, conferred by
Temozolomide: Mechanisms of Action, Repair and Resistance Current Molecular Pharmacology, 2012, Vol. 5, No. 1 107

multiple mechanisms, results in treatment failure. Intrinsic It has been corroborated in vivo that GBM MSH mutations,
glioma resistance may be a consequence of the presence selected during TMZ treatment correlate with TMZ
within tumours of multi-drug resistance proteins (MRP)1, resistance. MSH6 mutations, and/or absent protein
MRP3, MRP5, and glutathione-S-transferase (GST)  [81]. expression were neither found in pre-treatment GBM nor
It has recently confirmed that expression of MDR1/ABCB1 radiotherapy post-treatment GBM, but were detected in
encoding multi-drug resistant p-glycoprotein negatively approximately half of recurrent GBM patients treated with
impacts sensitivity to TMZ [82]. TMZ, a small lipophilic TMZ and radiotherapy, strongly indicating that MSH6
molecule is able to cross the blood brain barrier (BBB), an alterations in the tumour cell genome are associated with
obstacle to delivery of most agents to brain tumours. alkylating agent therapy and resistance. Indeed, MSH6
Furthermore, gliomas are naturally resistance to apoptosis as mutations have been found in 26% recurrent GBM cases
a consequence of phosphate and tensin homologue on following alkylating agent chemotherapy [93].
chromosome 10 (PTEN) tumour suppressor mutation and GBM patients possessing tumour MGMT promoter
constitutively active Akt/PI3K/mTOR/NF-kB signaling [83]. methylation respond well to alkylating agent therapy initially
Inherent resistance or tolerance to TMZ treatment may be [49], however, alkylating agent treatment of such MGMT-
conferred, as discussed, by MGMT activity, or MMR deficient GBM confers a strong selective pressure to lose
deficiency respectively. Thus adaptive alterations in DNA MMR function, resulting in O6-MeG tolerance and
repair pathways play critical roles in development of resistance to cell death [94]. Recent comprehensive genomic
resistance to TMZ. Many studies have now demonstrated a characterisation of human GBM genes revealed that initial
robust inverse relationship between MGMT expression and methylation of the MGMT promoter in conjunction with
inherent sensitivity to TMZ in vivo and in vitro [48, 49, 84, alkylating agent treatment leads to a shift in the mutation
85] (Fig. 2). It has also been reported that acquired TMZ spectrum affecting mutations at MMR gene loci. Thus,
resistance attributed to enhanced MGMT activity arises patients who initially respond to therapy may evolve not
frequently in glioma cell lines and xenografts [86]. Zhang only treatment resistance but also an MMR-defective
and coworkers [87] report that up-regulation of MGMT hypermutator phenotype.
expression in U373 GBM cells exposed to incremental N-Me purine lesions are repaired by BER, a major
concentrations of TMZ confers > 4-fold resistance to TMZ contributor to cellular resistance to TMZ [95]. The
in this TMZ-acquired resistant cell line. MGMT is an importance of BER was confirmed in SNB19 human
inducible DNA repair gene which can be up-regulated by not glioblastoma cells generated to possess acquired resistance
only alkylating agents but also ionising radiation and to TMZ. In these cells, O6-MeG lesions were tolerated
glucocorticoids [88]. GBM MGMT activity has been following loss of MSH6 MMR protein. The PARP inhibitor
compared in newly diagnosed patients, and recurrent GBM NU1025 partially restored sensitivity to TMZ (3.5x) in these
patients receiving combined radio- and alkylating agent- cells. Moreover, transcription of NTHL1 gene, encoding a
(TMZ, chloroethylnitrosourea) therapy. MGMT activity in key BER protein, possessing both abasic endonuclease and
untreated tumour samples was 37 fmol/mg protein, but in N-glycosylase activity, was elevated > 5-fold [87]. Up-
recurrent GBM MGMT activity of 182 fmol/mg protein was regulation of APE-1 (Ref-1) is characteristic of human
determined [85], a consequence either of selection of gliomas, contributing to TMZ resistance [96]. APE-1 activity
MGMT-expressing cells, or induction of MGMT by
is also able to promote resistance to radiotherapy plus TMZ
alkylating agents.
or BCNU in medulloblastoma and primitive neuroectoderm
The relatively low incidence of MSI in high grade tumours [97]. Reduction in APE-1 protein and its
gliomas would suggest that MMR deficiency is rarely endonuclease activity using APE-1-directed antisense
involved in intrinsic resistance to alkylating agents in this oligonucleotides in SNB19 GBM cells has been shown to
disease [89]. However, a large body of data implicates MMR decrease resistance to TMZ and BCNU. Hence, APE-1 may
deficiency in development of tolerance to O6-MeG lesions, present both a predictive and prognostic marker for
and therefore resistance to TMZ [90, 91]. Somatic mutations alkylating agent treatment [98], and a potential therapeutic
harboured within MMR proteins MSH2, MLH1 and MSH6 target [99].
in glioma cell lines, GBM xenografts and patient tumour
samples after treatment with TMZ is associated with TMZ Elucidation of mechanisms of tumour resistance to TMZ
tolerance regardless of MGMT expression [87, 92, 93]. has helped steer therapeutic strategies attempting to
Analysis of a large number of clinical samples revealed loss overcome resistance. In vitro tissue culture studies are
of MSH6 protein in a subset of recurrent GBM patients integral to understanding molecular mechanisms responsible
treated with TMZ, and that this loss was associated with for resistance. Exposure of tumour cells to TMZ selects cell
progressive disease during TMZ therapy [90]. MSH6 protein populations with acquired resistance or tolerance to this
down-regulation was observed in SNB19 GBM cells agent, disrupting or usurping the same molecular
previously exposed to increasing concentrations of TMZ, mechanisms which underlie acquired resistance to TMZ
proliferating in medium spiked with 100 μM TMZ. These emerging clinically [85, 87, 90]. Comprehensive
cells demonstrated 7.8-fold acquired resistance to TMZ understanding of such molecular mechanisms may reveal
compared with their TMZ-naive parent cell line [87]. Studies novel targets for therapy, guide rational chemotherapy
undertaken by Yip et al. [93] confirmed that MSH6 combination regimens to modulate resistance, or lead to
inactivation in vitro results in increased resistance to TMZ; development of novel TMZ analogues to surmount
reconstitution of MSH6 expression restored TMZ sensitivity. resistance mechanisms.
108 Current Molecular Pharmacology, 2012, Vol. 5, No. 1 Zhang et al.

OVERCOMING RESISTANCE TO TMZ lomeguatrib / TMZ combination therapy compared with


TMZ treatment alone. However, myelosuppression remains a
Inhibition of MGMT prohibitive limiting side effect to the use of MGMT
inhibitors and alkylating agent combination cancer
As direct repair of O6-MeG adducts by MGMT protein chemotherapy. This is a consequence of low MGMT
has a major impact on alkylating agent resistance clinically, expression within bone marrow which renders this tissue
a number of therapeutic approaches have been explored to susceptible to cytotoxicity [109]. To protect haematopoeitic
modulate MGMT activity and enhance drug response [100]. cells during chemotherapy, the strategy of gene transfer of
The potent non-toxic inhibitors of MGMT O6-benzyl mutant MGMT cDNA, encoding protein resistant to
guanine (O6-BG) and O6-(4-bromothenyl)guanine inactivation has been developed [36, 110, 111]. A phase I
(lomeguatrib; PaTrin-2; Fig. 6), have been used in clinical clinical study of such myelosuppressive gene therapy is
trials to deplete MGMT before administration of alkylating underway in the U.S.
agent therapy [44, 101, 102]. O6-BG acts as a
pseudosubstrate and binds to MGMT, covalently transferring
the benzyl moiety to the active site cysteine residue 145, INHIBITION OF BER
causing its irreversible inactivation. O6-BG is not
incorporated into the DNA of living cells, reacting directly Poly(ADP-ribose)polymerase Inhibition
with both cytoplasmic and nuclear MGMT [102]. Pre- N-Me purines, rapidly repaired by BER, contribute little
treatment of tumour cells containing high MGMT levels with to TMZ-induced toxicity in the absence of MGMT and
O6-BG enhances TMZ activity in vitro and in vivo, but has presence of proficient MMR. However, when O6-MeG is
little effect on tumour cells possessing low or undetectable repaired (MGMT) or tolerated (MMR-), and BER is
MGMT levels [14]. disrupted by PARP inhibition, N7-MeG and N3-MeA
Lomeguatrib is an orally bioavailable potent contribute significantly to TMZ cytotoxicity [72, 112]. It was
pseudosubstrate for MGMT. Covalent transfer of the to test the hypothesis that PARP inhibition could potentiate
bromothenyl group to the active site cysteine inactivates TMZ activity that PARP inhibitor AG 014699 first entered
MGMT. Lomeguatrib has shown promising activity in clinical trials in cancer patients [113-116]. The combination
sensitising a variety of human tumour xenografts to the revealed increased response rates and median time to
growth-inhibitory effects of O6-alkylating agents, including progression compared with TMZ treatment alone. ABT 888
TMZ and 1,3-bis-(2-chloroethyl)-1-nitrosurea, at the expense demonstrated broad in vivo activity in combination with
of only limited additional toxicity [103, 104]. A Phase I TMZ in diverse tumours [117]. At least 8 PARP inhibitors
clinical trial combining lomeguatrib and TMZ [105] led to a are currently undergoing Phase I, II, or III clinical
randomised Phase II study in 100 patients with metastatic evaluation, either in combination with chemotherapeutic
melanoma [106]. In this study, the efficacy of combination agents or alone, for treatment of malignant solid tumours
treatment was similar to that of TMZ treatment alone in [112, 113, 117]. The chemical structures of three such small
terms of response rates and median time to disease molecule inhibitors of PARP-1: olaparib, ABT 888 and BSI-
progression. However, the lomeguatrib schedule adopted 201 are shown in Fig. (7). It was hypothesised that single
permitted rapid recovery of tumour MGMT within 24 h. agent PARP inhibitor therapy would provide a synthetic
Subsequent clinical trials established a lethal strategy to target cancers with specific DNA repair
pharmacodynamically effective schedule and doses of defects. Indeed, PARP inhibition demonstrates substantial
lomeguatrib which effects complete and consistent MGMT single agent antitumour activity, possessing a wide
depletion in melanoma, CNS, prostate and colorectal therapeutic index in homologous DNA repair-defective
tumours [107, 108]. Moreover, significantly higher levels of tumours such as those arising in BRCA1 and BRCA2
O6-MeG adducts were present in PBMC DNA following mutation carriers; responses to olaparib (KU-0059436;
AZD2281) have been observed in Phase I and II studies

A B
O

N
N

H2N N N
H

Fig. (6). Structures of A) O6-benzylguanine and B) lomeguatrib.


Temozolomide: Mechanisms of Action, Repair and Resistance Current Molecular Pharmacology, 2012, Vol. 5, No. 1 109

O
A F
N

O
N B
NH H2N O

O
N H3C

N HN
H2N O H
C

NO2

Fig. (7). Structures of PARP inhibitors A) olaparib, B) ABT 888 and C) BSI-201 under clinical evaluation.

[117-119]. PARP inhibitor efficacy is also being assessed for DESIGN OF IMIDAZOTETRAZINE ANALOGUES TO
treatment of triple negative metastatic breast, and PTEN CIRCUMVENT MGMT REPAIR
mutant tumours [120].
The TMZ molecule acts as a methyl group delivery
Inhibiton of APE-1 vehicle imparting its toxic lode on O6-G. Primary inherent
TMZ resistance, conferred by MGMT, leads to
Pharmacological inhibition of BER using PARP stoichiometric removal of O6-methyl adducts restoring
inhibitors either alone or in combination with chemotherapy guanine. Logically, one could postulate that synthesis of an
such as TMZ has shown promise in clinical trials. However, imidazotetrazine analogue might be possible, that would
acquired resistance to PARP inhibitors is inevitably deliver an O6-G adduct not recognised by MGMT. In vitro
beginning to emerge; indeed up-regulation of homologous results of such synthetic efforts are summarised in Table 1
recombination repair to compensate for diminished BER has [60].
been identified [121]. Obligatory intermediates of BER are
Imidazotetrazine (series A) TMZ and its ring-opened
potentially cytotoxic AP sites, processed by APE-1. Abbots
triazene (series B) MTIC reveal growth inhibitory activity
and Madhusudan [122] highlight data that validate APE-1 as
an anticancer target. Indeed, small molecule inhibitors of (GI50 < 100 μM) against vector control GBM cell lines
SNB19V and U373V; however, activity is abrogated in their
APE-1 are able to potentiate alkylating agent cytotoxicty in
MGMT transfected SNB19M and U373M isogenic partners.
preclincial models. The topoisomerase II inhibitor
Compounds possessing a large lipophilic R group (1) and (2)
lucanthrone also inhibits APE-1, causing accumulation of
are much less active, whereas the trimethylsilymethyl
AP sites [123] potentiating TMZ cytotoxicty [124].
derivative (3) shows a pattern of activity similar to that of
Methoxyamine (MA) binds directly to AP sites and TMZ.
indirectly inhibits APE-1 by preventing AP site processing
Compounds with a small more polar group, e.g.
by APE-1; thus, AP sites accumulate. MA potentiates the
chloromethyl (5) or MOM (6) are more potent than TMZ
cytotoxicty of a wide variety of DNA damaging agents in
against vector control cell lines and also retain their activity
vitro and in xenograft studies [124-126] in both MMR
against isogenic MGMT overexpressing variants. Similarly
proficient and deficient tumour cells. A phase 1 clinical
study combining MA with TMZ in advanced solid tumours the imidazotetrazine esters (7 and 9) demonstrate a “flat”
distribution of activity across the four GBM cell lines; ring-
is currently underway.
opened triazene counterparts (8 and 10) are approximately
APE-1 is a multifunctional enzyme possessing a DNA equiactive to their cyclic imidazotetrazine precursors. These
repair domain and a redox domain. It is often referred to as results imply that the new imidazotetrazine TMZ analogues
Ref-1 and is involved in redox-sensitive activation of are ring-opened to create DNA alkylating species which
transcription factors including P53, NFkB, AP1 and HIF-1. generate cytotoxic lesions irreparable by MGMT. However,
Thus, synthesis of small molecules which directly target in vivo, imidazotetrazine esters would be substrates for
APE-1 may hold wide therapeutic benefit in the cancer plasma esterases, and the corresponding imidazotetrazine
arena. carboxylic acid 11 of methyl ester 7, for example reveals
110 Current Molecular Pharmacology, 2012, Vol. 5, No. 1 Zhang et al.

Table 1. Effect of Imidazotetrazine Anlogues (Series A and B) on GBM Cell Growth

H2NOC
N
N H2NOC
N H
N N N N
R N R
N
O NH
A B

Cell Lines GI50 (μM)


Compound Series R R
SNB19V SNB19M U373V U373M

TMZ A Me Me 35.7 ± 12.0 469.9 ± 88.7 68.0 ± 32.0 368.7 ± 86.1


MTIC B Me Me 50.9 ± 17.9 472.1 ± 88.7 93.7 ± 39.7 397.9 ± 33.7
1 A CH2Ph CH2Ph 140.5 ± 79.8 180.0 ± 86.3 165.0 ± 107.8 177.7 ± 133.7
2 A CH2CH2 Ph CH2CH2 Ph 98.6 ± 26.2 85.9 ± 13.7 114.7 ± 91.0 91.1 ± 38.3
3 A CH2Si(Me) 3 CH2Si(Me) 3 28.4 ± 5.9 302.4 ± 69.3 45.6 ± 14.5 249.5 ± 72.1
4 A CH2CF3 CH2CF3 39.9 ± 5.9 52.4 ± 11.5 102.5 ± 32.9 94.5 ± 24.0
5 A CH2Cl CH2Cl 34.6 ± 14.1 31.0± 7.9 29.3 ± 7.8 24.4 ± 1.0
6 A CH2OMe CH2OMe 30.8 ± 9.0 32.9 ± 9.2 26.0 ± 5.0 33.1 ± 9.3
7 A CH2CO2 Me CH2CO2 Me 47.5 ± 17.5 49.0 ± 14.7 32.1 ± 21.9 46.4 ± 16.7
8 B CH2CO2 Me CH2CO2 Me 47.7 ± 6.1 49.7 ± 7.3 32.6 ± 20.5 46.4 ± 9.8
9 A CH2CO2 Et CH2CO2 Et 52.4 ± 4.5 61.1 ± 5.2 63.2 ± 10.7 55.6 ± 6.5
10 B CH2CO2 Et CH2CO2 Et 33.6 ± 2.6 37.4 ± 11.9 30.3 ± 20.4 23.4 ± 12.2
11 A CH2CO2H CH2CO2H 339.7 ± 130.5 297.7 ± 68.4 269.0 ± 205.0 198.0± 91.1

poor activity against vector control and MGMT transfected ABBREVIATIONS


SNB19 and U373 GBM cell lines (GI50 > 195 μM).
ABCB1 = ATP-binding cassette protein B1
CONCLUDING REMARKS ADPRT = ADP-ribosyltransferase
AGT = alkylguanine DNA alkyltransferase
TMZ offers hope to newly diagnosed GBM patients.
However, in tumours expressing MGMT, the primary AIC = 5-aminoimidazole-4-carboxamide
cytotoxic lesion is stoichiometrically removed. These
AP = abasic
tumours are inherently resistant to TMZ.
APE-1 = AP endonuclease-1
While potentiation of TMZ activity can be conferred by
agents that deplete MGMT, adverse, therapy-limiting BBB = blood brain barrier
complications emerge. The majority of lesions induced by BCNU = 1,3-bis-(2-chloroethyl)-1-nitrosourea
TMZ are subject to rapid BER; interruption of BER by (Carmustine)
PARP inhibition is also able to potentiate TMZ response.
However, in tumours initially responsive to TMZ therapy, BER = base excision repair
acquired resistance or tolerance to treatment evolves, tumour CNS = central nervous system
cells cunningly adapt and modify DNA repair machinery,
MGMT and BER proteins are up-regulated, MMR is DSB = DNA double strand break
disabled, exposing DNA, oblivious to TMZ-induced lesions, dRp = 5`-deoxyribose phosphate
vulnerable to further mutagenic events promoting a more
resistant and aggressive GBM cancer cell. It is thus evident GBM = glioblastoma multiforme
that formidable therapeutic challenges persist, and GST = glutathione-S-transferase
development of novel agents is vital to augment current
HNPCC = hereditary nonpolyposis colorectal cancer
treatment of this intractable disease.
MDR = multidrug resistance
ACKNOWLEDGEMENTS MGMT = methylguanine DNA methyltransferase
We thank Marc Hummersone and the chemists of MMR = DNA mismatch repair
Pharminox Ltd synthesis of novel imidazotetrazine and
MRP = multidrug resistance protein
imidazotriazene molecules.
Temozolomide: Mechanisms of Action, Repair and Resistance Current Molecular Pharmacology, 2012, Vol. 5, No. 1 111

MSI = microsatellite instability positron emission tomography. Ann. Neurol., 1984, (Suppl. 15),
S98-102.
MTIC = 5-(3-methyltriazen-1-yl)-imidazole-4- [11] Drabløs, F.; Feyzi, E.; Aas, P.A.; Vaagbø, C.B.; Kavli, B.; Bratlie,
carboxamide M.S.; Peña-Diaz, J.; Otterlei, M.; Slupphaug, G.; Krokan, H.E.
Alkylation damage in DNA and RNA--repair mechanisms and
NAD+ = -nicotinamide adenine dinucleotide medical significance. DNA Repair, 2004, 3(11), 1389-1407.
[12] Wedge, S.R.; Porteous, J.K.; Newlands, E.S. 3-aminobenzamide
N3-MeA = N3-methyladenine and/or O6-benzylguanine evaluated as an adjuvant to
N7-MeG = N7-methylguanine temozolomide or BCNU treatment in cell lines of variable
mismatch repair status and O6-alkylguanine-DNA alkyltransferase
O6-MeG = O6-methylguanine activity. Br. J. Cancer, 1996, 74(7), 1030-1036.
[13] Wedge, S.R.; Newlands, E.S. O6-benzylguanine enhances the
PARP-1 = poly(ADP-ribose)polymerase-1 sensitivity of a glioma xenograft with low O6-alkylguanine-DNA
alkyltransferase activity to temozolomide and BCNU. Br. J.
PTEN = phosphatise and tensin homologue Cancer, 1996, 73(9), 1049-1052.
SSB = DNA single strand break [14] Wedge, S.R.; Porteus, J.K.; May, B.L.; Newlands, E.S. Potentiation
of temozolomide and BCNU cytotoxicity by O(6)-benzylguanine: a
TMZ = temozolomide comparative study in vitro. Br. J. Cancer, 1996, 73(4), 482-490.
[15] Kyrtopoulos, S.A.; Anderson, L.M.; Chhabra, S.K.; Souliotis, V.L.;
XRCC1 = X-ray repair cross-complementing 1 Pletsa, V.; Valavanis, C.; Georgiadis, P. DNA adducts and the
mechanism of carcinogenesis and cytotoxicity of methylating
agents of environmental and clinical significance. Cancer Detect.
REFERENCES
Prev., 1997, 21(5), 391-405.
[16] Margison, G.P.; Santibanez-Koref, M.F. O-6-alkylguanine-DNA
[1] Laperriere, N.; Zuraw, L.; Cairncross, G.; Cancer Care Ontario alkyltransferase: role in carcinogenesis and chemotherapy.
Practice Guidelines Initiative Neuro-Oncology Disease Site Group. Bioessays, 2002, 24(3), 255-266.
Radiotherapy for newly diagnosed malignant glioma in adults: a [17] Mojas, N.; Lopes, M.; Jiricny, J. Mismatch repair-dependent
systematic review. Radiother. Oncol., 2002, 64(3), 259-273. processing of methylation damage gives rise to persistent single-
[2] Huse, J.T.; Holland, E.C. Targeting brain cancer: advances in the stranded gaps in newly replicated DNA. Genes Dev., 2007, 21(24),
molecular pathology of malignant glioma and medulloblastoma. 3342-3355.
Nat. Rev. Cancer, 2010, 10(5), 319-331. [18] Cejka, P.; Stojic, L.; Mojas, N.; Russell, A.M.; Heinimann, K.;
[3] Furnari, F.B.; Fenton, T.; Bachoo, R.M.; Mukasa, A.; Stommel, Cannavo, E.; di Pietro, M.; Marra, G.; Jiricny, J. Methylation-
J.M.; Stegh, A.; Hahn, W.C.; Ligon, K.L.; Louis, D.N.; Brennan, induced G(2)/M arrest requires a full complement of the mismatch
C.; Chin, L.; DePinho, R.A.; Cavenee, W.K. Malignant astrocytic repair protein hMLH1. EMBO J., 2003, 22(9), 2245-2254.
glioma: genetics, biology, and paths to treatment. Genes Dev., [19] Roos, W.; Baumgartner, M.; Kaina, B. Apoptosis triggered by
2007, 21(21), 2683-2710. DNA damage O-6-methylguanine in human lymphocytes requires
[4] Stupp, R.; Hegi, M.E.; Mason, W.P.; van den Bent, M.J.; DNA replication and is mediated by p53 and Fas/CD95/Apo-1.
Taphoorn, M.J.B.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, Oncogene, 2004, 23(2), 359-367.
B.; Belanger, K.; Hau, P.; Brandes, A.A.; Gijtenbeek, J.; Marosi, [20] Zhukovskaya, N.; Branch, P.; Aquilina, G.; Karran, P. DNA
C.; Vecht, C.J.; Mokhtari, K.; Wesseling, P.; Villa, S.; Eisenhauer, replication arrest and tolerance to DNA methylation damage.
E.; Gorlia, T.; Weller, M.; Lacombe, D.; Cairncross, J.G.; Carcinogenesis, 1994, 15(10), 2189-2194.
Mirimanoff, R.O.; European Organisation for Research and [21] Stojic, L.; Brun, R.; Jiricny, J. Mismatch repair and DNA damage
Treatment of Cancer Brain Tumour and Radiation Oncology signalling. DNA Repair (Amst), 2004, 3(8-9), 1091-1101.
Groups; National Cancer Institute of Canada Clinical Trials Group. [22] D'Atri, S.; Tentori, L.; Lacal, P.M.; Graziani, G.; Pagani, E.;
Effects of radiotherapy with concomitant and adjuvant Benincasa, E.; Zambruno, G.; Bonmassar, E.; Jiricny, J.
temozolomide versus radiotherapy alone on survival in Involvement of the mismatch repair system in temozolomide-
glioblastoma in a randomised phase III study: 5-year analysis of the induced apoptosis. Mol. Pharmacol., 1998, 54(2), 334-341.
EORTC-NCIC trial. Lancet Oncol., 2009, 10(5), 459-466. [23] Horton, J.K.; Wilson, S.H. Hypersensitivity phenotypes associated
[5] Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, with genetic and synthetic inhibitor-induced base excision repair
B.; Taphoorn, M.J.; Belanger, K.; Brandes, A.A.; Marosi, C.; deficiency. DNA Repair, 2007, 6(4), 530-543.
Bogdahn, U.; Curschmann, J.; Janzer, R.C.; Ludwin, S.K.; Gorlia, [24] Lim, A.; Li, B.F. The nuclear targeting and nuclear retention
T.; Allgeier, A.; Lacombe, D.; Cairncross, J.G.; Eisenhauer, E.; properties of a human DNA repair protein O6-methylguanine-DNA
Mirimanoff, R.O.; European Organisation for Research and methyltransferase are both required for its nuclear localization: the
Treatment of Cancer Brain Tumor and Radiotherapy Groups; possible implications. EMBO J., 1996, 15(15), 4050-4060.
National Cancer Institute of Canada Clinical Trials Group. [25] Coulter, R.; Blandino, M.; Tomlinson, J.M.; Pauly, G.T.;
Radiotherapy plus concomitant and adjuvant temozolomide for Krajewska, M.; Moschel, R.C.; Peterson, L.A.; Pegg, A.E.; Spratt,
glioblastoma. N. Engl. J. Med., 2005, 352(10), 987-996. T.E. Differences in the rate of repair of O6-alkylguanines in
[6] Newlands, E.S.; Blackledge, G.R.; Slack, J.A.; Rustin, G.J.; Smith, different sequence contexts by O6-alkylguanine-DNA
D.B.; Stuart, N.S.; Quarterman, C.P.; Hoffman, R.; Stevens, M.F.; alkyltransferase. Chem. Res. Toxicol., 2007, 20(12), 1966-1971.
Brampton, M.H. Phase I trial of temozolomide (CCRG 81045: [26] Fang, Q.; Noronha, A.M.; Murphy, S.P.; Wilds, C.J.; Tubbs, J.L.;
M&B 39831: NSC 362856). Br. J. Cancer, 1992, 65(2), 287-291. Tainer, J.A.; Chowdhury, G.; Guengerich, F.P.; Pegg, A.E. Repair
[7] Tsang, L.L.; Farmer, P.B.; Gescher, A.; Slack, J.A. of O6-G-alkyl-O6-G interstrand cross-links by human O6-
Characterisation of urinary metabolites of temozolomide in humans alkylguanine-DNA alkyltransferase. Biochemistry, 2008, 47(41),
and mice and evaluation of their cytotoxicity. Cancer Chemother. 10892-10903.
Pharmacol., 1990, 26(6), 429-436. [27] Pegg, A.E. Repair of O(6)-alkylguanine by alkyltransferases.
[8] Denny, B.J.; Wheelhouse, R.T.; Stevens, M.F.; Tsang, L.L.; Slack, Mutat. Res., 2000, 462(2-3), 83-100.
J.A. NMR and molecular modeling investigation of the mechanism [28] Pegg, A.E.; Dolan, M.E.; Moschel, R.C. Structure, function, and
of activation of the antitumor drug temozolomide and its inhibition of O6-alkylguanine-DNA alkyltransferase. Prog. Nucleic
interaction with DNA. Biochemistry, 1994, 33, 9045- 9051. Acid Res. Mol. Biol., 1995, 51, 167-223.
[9] Tisdale, M.J. Antitumor imidazotetrazines--XV. Role of guanine [29] Srivenugopal, K.S.; Yuan, X.H.; Friedman, H.S.; Ali-Osman, F.
O6 alkylation in the mechanism of cytotoxicity of Ubiquitination-dependent proteolysis of O6-methylguanine-DNA
imidazotetrazinones. Biochem. Pharmacol., 1987, 36(4), 457-462. methyltransferase in human and murine tumor cells following
[10] Rottenberg, D.A.; Ginos, J.Z.; Kearfott, K.J.; Junck, L.; Bigner, inactivation with O6-benzylguanine or 1,3-bis(2-chloroethyl)-1-
D.D. In vivo measurement of regional brain tissue pH using nitrosourea. Biochemistry, 1996, 35(4), 1328-1334.
112 Current Molecular Pharmacology, 2012, Vol. 5, No. 1 Zhang et al.

[30] Kaina, B.; Christmann, M. DNA repair in resistance to alkylating Methylguanine-DNA Methyltransferase Promoter Methylation in
anticancer drugs. Int .J. Clin. Pharmacol. Ther., 2002, 40(8), 354- Glioblastoma Patients Treated with Temozolomide. Clin. Cancer
367. Res., 2004, 10(6), 1871-1874.
[31] Margison, G.P.; Povey, A.C.; Kaina, B.; Santibanez Koref, M.F. [49] Hegi, M.E.; Liu, L.; Herman, J.G.; Stupp, R.; Wick, W.; Weller,
Variability and regulation of O6-alkylguanine-DNA M.; Mehta, M.P.; Gilbert, M.R. Correlation of O6-Methylguanine
alkyltransferase. Carcinogenesis, 2003, 24(4), 625-635. Methyltransferase (MGMT) Promoter Methylation With Clinical
[32] Kaina, B.; Christmann, M.; Naumann, S.; Roos, W.P. MGMT: Key Outcomes in Glioblastoma and Clinical Strategies to Modulate
node in the battle against genotoxicity, carcinogenicity and MGMT Activity. J. Clin. Oncol., 2008, 26(25), 4189-4199.
apoptosis induced by alkylating agents. DNA Repair, 2007, 6(8), [50] Bignami, M.; Casorelli, I.; Karran, P. Mismatch repair and
1079-1099. response to DNA-damaging antitumour therapies. Eur. J. Cancer,
[33] Mouawad, R.; Sebert, M.; Michels, J.; Bloch, J.; Spano, J.P.; 2003, 39(15), 2142-2149.
Khayat, D. Treatment for metastatic malignant melanoma: old [51] Christmann, M.; Tomicic, M.T.; Roos, W.P.; Kaina, B.
drugs and new strategies. Crit. Rev. Oncol. Hematol., 2010, 74(1), Mechanisms of human DNA repair: an update. Toxicology, 2003,
27-39. 193(1-2), 3-34.
[34] Silber, J.R.; Blank, A.; Bobola, M.S.; Ghatan, S.; Kolstoe, D.D.; [52] Umar, A.; Risinger, J.I.; Glaab, W.E.; Tindall, K.R.; Barrett, J.C.;
Berger, M.S. O6-methylguanine-DNA methyltransferase-deficient Kunkel, T.A. Functional overlap in mismatch repair by human
phenotype in human gliomas: frequency and time to tumor MSH3 and MSH6. Genetics, 1998, 148(4), 1637-1646.
progression after alkylating agent-based chemotherapy. Clin. [53] Chai, S.M.; Zeps, N.; Shearwood, A.M.; Grieu, F.; Charles, A.;
Cancer Res., 1999, 5(4), 807-814. Harvey, J.; Goldblatt, J.; Joseph, D.; Iacopetta, B. Screening for
[35] Gerson, S.L. Clinical relevance of MGMT in the treatment of defective DNA mismatch repair in stage II and III colorectal cancer
cancer. J. Clin. Oncol., 2002, 20(9), 2388-2399. patients. Clin. Gastroenterol. Hepatol., 2004, 2(11), 1017-1025.
[36] Gerson, S.L. MGMT: its role in cancer aetiology and cancer [54] Gologan, A.; Sepulveda, A.R. Microsatellite instability and DNA
therapeutics. Nat. Rev. Cancer, 2004, 4(4), 296-307. mismatch repair deficiency testing in hereditary and sporadic
[37] Gerson, S.L.; Willson, J.K. O6-alkylguanine-DNA alkyltransferase. gastrointestinal cancers. Clin. Lab. Med., 2005, 25(1), 179-196.
A target for the modulation of drug resistance. Hematol. Oncol. [55] Peltomaki, P. Role of DNA Mismatch Repair Defects in the
Clin. North Am., 1995, 9(2), 431-450. Pathogenesis of Human Cancer. J. Clin. Oncol., 2003, 21(6), 1174-
[38] Rolhion, C.; Penault-Llorca, F.; Kemeny, J.L.; Kwiatkowski, F.; 1179.
Lemaire, J.J.; Chollet, P.; Finat-Duclos, F.; Verrelle, P. O(6)- [56] Fink, D.; Aebi, S.; Howell, S.B. The role of DNA mismatch repair
methylguanine-DNA methyltransferase gene (MGMT) expression in drug resistance. Clin. Cancer Res., 1998, 4(1), 1-6.
in human glioblastomas in relation to patient characteristics and [57] Grady, W.M.; Carethers, J.M. Genomic and epigenetic instability
p53 accumulation. Int. J. Cancer, 1999, 84(4), 416-420. in colorectal cancer pathogenesis. Gastroenterology, 2008, 135(4),
[39] Bacolod, M.D.; Johnson, S.P.; Pegg, A.E.; Dolan, M.E.; Moschel, 1079-1099.
R.C.; Bullock, N.S.; Fang, Q.; Colvin, O.M.; Modrich, P.; Bigner, [58] Herman, J.G.; Umar, A.; Polyak, K.; Graff, J.R.; Ahuja, N.; Issa,
D.D.; Friedman, H.S. Brain tumor cell lines resistant to O6- J.P.; Markowitz, S.; Willson, J.K.; Hamilton, S.R.; Kinzler, K.W.;
benzylguanine/1,3-bis(2-chloroethyl)-1-nitrosourea chemotherapy Kane, M.F.; Kolodner, R.D.; Vogelstein, B.; Kunkel, T.A. Baylin,
have O6-alkylguanine-DNA alkyltransferase mutations. Mol. S.B. Incidence and functional consequences of hMLH1 promoter
Cancer Ther., 2004, 3(9), 1127-1135. hypermethylation in colorectal carcinoma. Proc. Natl. Acad. Sci.
[40] Esteller, M. Aberrant DNA methylation as a cancer-inducing USA, 1998, 95(12), 6870-6875.
mechanism. Annu. Rev. Pharmacol. Toxicol., 2005, 45, 629-656. [59] Grady, W.M.; Rajput, A.; Lutterbaugh, J.D.; Markowitz, S.D.
[41] Esteller, M.; Herman, J.G. Generating mutations but providing Detection of aberrantly methylated hMLH1 promoter DNA in the
chemosensitivity: the role of O6-methylguanine DNA serum of patients with microsatellite unstable colon cancer. Cancer
methyltransferase in human cancer. Oncogene, 2004, 23(1), 1-8. Res., 2001, 61(3), 900-902.
[42] Middleton, M.R.; Margison, G.P. Improvement of chemotherapy [60] Zhang, J.S.M.; Hummersone, M.; Madhusudan, S.; Laughton, CA.;
efficacy by inactivation of a DNA-repair pathway. Lancet Oncol., Bradshaw, T.D. Certain imidazotetrazines escape O6-
2003, 4(1), 37-44. methylguanine-DNA methyltransferase and mismatch repair
[43] Esteller, M.; Garcia-Foncillas, J.; Andion, E.; Goodman, S.N.; Oncology, 2011, in press.
Hidalgo, O.F.; Vanaclocha, V.; Baylin, S.B.; Herman, J.G. [61] Karran, P. Mechanisms of tolerance to DNA damaging therapeutic
Inactivation of the DNA-repair gene MGMT and the clinical drugs. Carcinogenesis, 2001, 22(12), 1931-1937.
response of gliomas to alkylating agents. N. Engl. J. Med., 2000, [62] Wood, R.D.; Mitchell, M.; Sgouros, J.; Lindahl, T. Human DNA
343(19), 1350-1354. repair genes. Science, 2001, 291(5507), 1284-1289.
[44] Sabharwal, A.; Middleton, M.R. Exploiting the role of O6- [63] Hickson, I.D., Ed. Base excision repair of DNA damage; Landes
methylguanine-DNA-methyltransferase (MGMT) in cancer Bioscience and Chapman and Hall: Austin, Texas, 1997. ISBN
therapy. Curr. Opin. Pharmacol., 2006, 6(4), 355-363. 0412131617.
[45] Blanc, J.L.; Wager, M.; Guilhot, J.; Kusy, S.; Bataille, B.; [64] Lindahl, T.; Karran, P.; Wood, R.D. DNA excision repair
Chantereau, T.; Lapierre, F.; Larsen, C.J.; Karayan-Tapon, L. pathways. Curr. Opin. Genet. Dev., 1997, 7(2), 158-169.
Correlation of clinical features and methylation status of MGMT [65] Dantzer, F.; Ame, J.C.; Schreiber, V.; Nakamura, J.; Menissier-de
gene promoter in glioblastomas. J. Neurooncol., 2004, 68(3), 275- Murcia, J.; de Murcia, G. Poly(ADP-ribose) polymerase-1
283. activation during DNA damage and repair. Methods Enzymol.,
[46] Stupp, R.; Hegi, M.E.; van den Bent, M.J.; Mason, W.P.; Weller, 2006, 409, 493-510.
M.; Mirimanoff, R.O.; Cairncross, J.G.; European Organisation for [66] Malanga, M.; Althaus, F.R. The role of poly(ADP-ribose) in the
Research and Treatment of Cancer Brain Tumor and Radiotherapy DNA damage signaling network. Biochem. Cell Biol., 2005, 83(3),
Groups; National Cancer Institute of Canada Clinical Trials Group. 354-364.
Changing paradigms - An update on the multidisciplinary [67] Aguilar-Quesada, R.; Munoz-Gamez, J.A.; Martin-Oliva, D.;
management of malignant glioma. Oncologist, 2006, 11(2), 165- Peralta-Leal, A.; Quiles-Perez, R.; Rodriguez-Vargas, J.M.; Ruiz
180. de Almodovar, M.; Conde, C.; Ruiz-Extremera, A.; Oliver, F.J.
[47] Hegi, M.E.; Diserens, A.; Gorlia, T.; Hamou, M.; de Tribolet, N.; Modulation of transcription by PARP-1: consequences in
Weller, M.; Kros, J.M.; Hainfellner, J.A.; Mason, W.; Mariani, L.; carcinogenesis and inflammation. Curr. Med. Chem., 2007, 14(11),
Bromberg, J.E.C.; Hau, P.; Mirimanoff, R.O.; Cairncross, J.G.; 1179-1187.
Janzer, R.C.; Stupp, R. MGMT gene silencing and benefit from [68] Lockett, K.L.; Snowhite, I.V.; Hu, J.J. Nucleotide-excision repair
temozolomide in glioblastoma. N. Engl. J. Med., 2005, 352(10), and prostate cancer risk. Cancer Lett., 2005, 220(2), 125-135.
997-1003. [69] Sobol, R.W.; Horton, J.K.; Kuhn, R.; Gu, H.; Singhal, R.K.;
[48] Hegi, M.E.; Diserens, A.-C.; Godard, S.; Dietrich, P.-Y.; Regli, L.; Prasad, R.; Rajewsky, K.; Wilson, S.H. Requirement of
Ostermann, S.; Otten, P.; Van Melle, G.; de Tribolet, N.; Stupp, R. mammalian DNA polymerase-beta in base-excision repair. Nature,
Clinical Trial Substantiates the Predictive Value of O-6- 1996, 379(6561), 183-186.
Temozolomide: Mechanisms of Action, Repair and Resistance Current Molecular Pharmacology, 2012, Vol. 5, No. 1 113

[70] Curtin, N.J.; Wang, L.Z.; Yiakouvaki, A.; Kyle, S.; Arris, C.A.; [88] Grombacher, T.; Mitra, S.; Kaina, B. Induction of the
Canan-Koch, S.; Webber, S.E.; Durkacz, B.W.; Calvert, H.A.; alkyltransferase (MGMT) gene by DNA damaging agents and the
Hostomsky, Z.; Newell, D.R. Novel poly(ADP-ribose) polymerase- glucocorticoid dexamethasone and comparison with the response of
1 inhibitor, AG14361, restores sensitivity to temozolomide in base excision repair genes. Carcinogenesis, 1996, 17(11), 2329-
mismatch repair-deficient cells. Clin. Cancer Res., 2004, 10(3), 2336.
881-889. [89] Eckert, A.; Kloor, M.; Giersch, A.; Ahmadi, R.; Herold-Mende, C.;
[71] Ratnam, K.; Low, J.A. Current development of clinical inhibitors Hampl, J.A.; Heppner, F.L.; Zoubaa, S.; Holinski-Feder, E.;
of poly(ADP-ribose) polymerase in oncology. Clin. Cancer Res., Pietsch, T.; Wiestler, O.D.; von Knebel Doeberitz, M.; Roth, W.;
2007, 13(5), 1383-1388. Gebert, J. Microsatellite instability in pediatric and adult high-
[72] Tentori, L.; Portarena, I.; Vernole, P.; Gold, B.; Graziani, G. grade gliomas. Brain Pathol., 2007, 17(2), 146-150.
Apoptotic and genotoxic effects of a methyl sulfonate ester that [90] Cahill, D.P.; Levine, K.K.; Betensky, R.A.; Codd, P.J.; Romany,
selectively generates N3-methyladenine and poly(ADP-ribose) C.A.; Reavie, L.B.; Batchelor, T.T.; Futreal, P.A.; Stratton, M.R.;
polymerase inhibitors in normal peripheral blood lymphocytes. Curry, W.T.; Iafrate, A.J.; Louis, D.N. Loss of the mismatch repair
Cancer Chemother. Pharmacol., 2002, 49(3), 217-224. protein MSH6 in human glioblastomas is associated with tumor
[73] Tentori, L.; Graziani, G. Recent approaches to improve the progression during temozolomide treatment. Clin. Cancer Res.,
antitumor efficacy of temozolomide. Curr. Med. Chem., 2009, 2007, 13(7), 2038-2045.
16(2), 245-257. [91] Cahill, D.P.; Codd, P.J.; Batchelor, T.T.; Curry, W.T.; Louis, D.N.
[74] Alvino, E.; Castiglia, D.; Caporali, S.; Pepponi, R.; Caporaso, P.; MSH6 inactivation and emergent temozolomide resistance in
Lacal, P.M.; Marra, G.; Fischer, F.; Zambruno, G.; Bonmassar, E.; human glioblastomas. Clin. Neurosurg., 2008, 55, 165-171.
Jiricny, J.; D'Atri, S. A single cycle of treatment with [92] Friedman, H.S.; Johnson, S.P.; Dong, Q.; Schold, S.C.; Rasheed,
temozolomide, alone or combined with O-6-benzylguanine, B.K.; Bigner, S.H.; Ali-Osman, F.; Dolan, E.; Colvin, O.M.;
induces strong chemoresistance in melanoma cell clones in vitro: Houghton, P.; Germain, G.; Drummond, J.T.; Keir, S.; Marcelli, S.;
Role of O-6-methylguanine-DNA methyltransferase and the Bigner, D.D.; Modrich, P. Methylator resistance mediated by
mismatch repair system. Int. J. Oncol., 2006, 29(4), 785-797. mismatch repair deficiency in a glioblastoma multiforme xenograft.
[75] Dean, M.; Fojo, T.; Bates, S. Tumour stem cells and drug Cancer Res., 1997, 57(14), 2933-2936.
resistance. Nat. Rev. Cancer, 2005, 5(4), 275-284. [93] Yip, S.; Miao, J.; Cahill, D.P.; Iafrate, A.J.; Aldape, K.; Nutt, C.L.;
[76] Singh, S.K.; Hawkins, C.; Clarke, I.D.; Squire, J.A.; Bayani, J.; Louis, D.N. MSH6 Mutations Arise in Glioblastomas during
Hide, T.; Henkelman, R.M.; Cusimano, M.D.; Dirks, P.B. Temozolomide Therapy and Mediate Temozolomide Resistance.
Identification of human brain tumour initiating cells. Nature, 2004, Clin. Cancer Res., 2009, 15(14), 4622-4629.
432(7015), 396-401. [94] Casorelli, I.; Russo, M.T.; Bignami, M. Role of mismatch repair
[77] Persidis, A. Cancer multidrug resistance. Nat. Biotechnol., 1999, and MGMT in response to anticancer therapies. Anticancer Agents
17(1), 94-95. Med. Chem., 2008, 8(4), 368-380.
[78] Bradshaw, T.D.; Stone, E.L.; Trapani, V.; Leong, C.O.; Matthews, [95] Trivedi, R.N.; Almeida, K.H.; Fornsaglio, J.L.; Schamus, S.; Sobol,
C.S.; te Poele, R.; Stevens, M.F. Mechanisms of acquired R.W. The role of base excision repair in the sensitivity and
resistance to 2-(4-Amino-3-methylphenyl)benzothiazole in breast resistance to temozolomide-mediated cell death. Cancer Res.,
cancer cell lines. Breast Cancer Res. Treat., 2008, 110(1), 57-68. 2005, 65(14), 6394-6400.
[79] Longley, D.B.; Johnston, P.G. Molecular mechanisms of drug [96] Silber, J.R.; Bobola, M.S.; Blank, A.; Schoeler, K.D.; Haroldson,
resistance. J. Pathol., 2005, 205(2), 275-292. P.D.; Huynh, M.B.; Kolstoe, D.D. The Apurinic/Apyrimidinic
[80] Shi, X.; Liu, S.; Kleeff, J.; Friess, H.; Buchler, M.W. Acquired Endonuclease Activity of Ape1/Ref-1 Contributes to Human
resistance of pancreatic cancer cells towards 5-Fluorouracil and Glioma Cell Resistance to Alkylating Agents and Is Elevated by
gemcitabine is associated with altered expression of apoptosis- Oxidative Stress. Clin. Cancer Res., 2002, 8(9), 3008-3018.
regulating genes. Oncology, 2002, 62(4), 354-362. [97] Bobola, M.S.; Finn, L.S.; Ellenbogen, R.G.; Geyer, J.R.; Berger,
[81] Calatozzolo, C.; Gelati, M.; Ciusani, E.; Sciacca, F.L.; Pollo, B.; M.S.; Braga, J.M.; Meade, E.H.; Gross, M.E.; Silber, J.R.
Cajola, L.; Marras, C.; Silvani, A.; Vitellaro-Zuccarello, L.; Croci, Apurinic/apyrimidinic endonuclease activity is associated with
D.; Boiardi, A.; Salmaggi, A. Expression of drug resistance response to radiation and chemotherapy in medulloblastoma and
proteins Pgp, MRP1, MRP3, MRP5 and GST-pi in human glioma. primitive neuroectodermal tumors. Clin. Cancer Res., 2005,
J. Neurooncol., 2005, 74(2), 113-121. 11(20), 7405-7414.
[82] Schaich, M.; Kestel, L.; Pfirrmann, M.; Robel, K.; Illmer, T.; [98] Bobola, M.S.; Emond, M.J.; Blank, A.; Meade, E.H.; Kolstoe,
Kramer, M.; Dill, C.; Ehninger, G.; Schackert, G.; Krex, D. A D.D.; Berger, M.S.; Rostomily, R.C.; Silbergeld, D.L.; Spence,
MDR1 (ABCB1) gene single nucleotide polymorphism predicts A.M.; Silber, J.R. Apurinic endonuclease activity in adult gliomas
outcome of temozolomide treatment in glioblastoma patients. Ann. and time to tumor progression after alkylating agent-based
Oncol., 2009, 20(1), 175-181. chemotherapy and after radiotherapy. Clin. Cancer Res., 2004,
[83] Lefranc, F.; Rynkowski, M.; DeWitte, O.; Kiss, R. Present and 10(23), 7875-7883.
potential future adjuvant issues in high-grade astrocytic glioma [99] Al-Attar, A.; Gossage, L.; Fareed, K.R.; Shehata, M.; Mohammed,
treatment. Adv. Tech. Stand. Neurosurg., 2009, 34, 3-35. M.; Zaitoun, A.M.; Soomro, I.; Lobo, D.N.; Abbotts, R.; Chan, S.;
[84] Bobola, M.S.; Tseng, S.H.; Blank, A.; Berger, M.S.; Silber, J.R. Madhusudan, S. Human apurinic/apyrimidinic endonuclease
Role of O6-methylguanine-DNA methyltransferase in resistance of (APE1) is a prognostic factor in ovarian, gastro-oesophageal and
human brain tumor cell lines to the clinically relevant methylating pancreatico-biliary cancers. Br. J. Cancer, 2010, 102(4), 704-709.
agents temozolomide and streptozotocin. Clin. Cancer Res., 1996, [100] Verbeek, B.; Southgate, T.D.; Gilham, D.E.; Margison, G.P. O6-
2(4), 735-741. Methylguanine-DNA methyltransferase inactivation and
[85] Wiewrodt, D.; Nagel, G.; Dreimuller, N.; Hundsberger, T.; chemotherapy. Br. Med. Bull., 2008, 85, 17-33.
Perneczky, A.; Kaina, B. MGMT in primary and recurrent human [101] Dolan, M.E.; Pegg, A.E. O6-benzylguanine and its role in
glioblastomas after radiation and chemotherapy and comparison chemotherapy. Clin. Cancer Res., 1997, 3(6), 837-847.
with p53 status and clinical outcome. Int. J. Cancer, 2008, 122(6), [102] Dolan, M.E.; Moschel, R.C.; Pegg, A.E. Depletion of mammalian
1391-1399. O6-alkylguanine-DNA alkyltransferase activity by O6-
[86] Kitange, G.J.; Carlson, B.L.; Schroeder, M.A.; Grogan, P.T.; benzylguanine provides a means to evaluate the role of this protein
Lamont, J.D.; Decker, P.A.; Wu, W.; James, C.D.; Sarkaria, J.N. in protection against carcinogenic and therapeutic alkylating
Induction of MGMT expression is associated with temozolomide agents. Proc. Natl. Acad. Sci. USA. 1990, 87(14), 5368-5372.
resistance in glioblastoma xenografts. Neuro. Oncol., 2009, 11(3), [103] Middleton, M.R.; Kelly, J.; Thatcher, N.; Donnelly, D.J.;
281-291. McElhinney, R.S.; McMurry, T.B.; McCormick, J.E.; Margison,
[87] Zhang, J.; Stevens, M.F.; Laughton, C.A.; Madhusudan, S.; G.P. O(6)-(4-bromothenyl)guanine improves the therapeutic index
Bradshaw, T.D. Acquired Resistance to Temozolomide in Glioma of temozolomide against A375M melanoma xenografts. Int. J.
Cell Lines: Molecular Mechanisms and Potential Translational Cancer, 2000, 85(2), 248-252.
Applications. Oncology, 2010, 78(2), 103-114.
114 Current Molecular Pharmacology, 2012, Vol. 5, No. 1 Zhang et al.

[104] Middleton, M.R.; Thatcher, N.; McMurry, T.B.; McElhinney, R.S.; [115] Palma, J.P.; Wang, Y.C.; Rodriguez, L.E.; Montgomery, D.; Ellis,
Donnelly, D.J.; Margison, G.P. Effect of O6-(4- P.A.; Bukofzer, G.; Niquette, A.; Liu, X.; Shi, Y.; Lasko, L.; Zhu,
bromothenyl)guanine on different temozolomide schedules in a G.D.; Penning, T.D.; Giranda, V.L.; Rosenberg, S.H.; Frost, D.J.;
human melanoma xenograft model. Int. J. Cancer, 2002, 100(5), Donawho, C.K. ABT-888 confers broad in vivo activity in
615-617. combination with temozolomide in diverse tumors. Clin. Cancer
[105] Ranson, M.; Middleton, M.R.; Bridgewater, J.; Lee, S.M.; Dawson, Res., 2009, 15(23), 7277-7290.
M.; Jowle, D.; Halbert, G.; Waller, S.; McGrath, H.; Gumbrell, L.; [116] Plummer, R.; Lorigan, P.; Evans, J.; Steven, N.; Middleton, M.;
McElhinney, R.S.; Donnelly, D.; McMurry, T.B.; Margison, G.P. Wilson, R.; Snow, K.; Dewji, R.; Calvert, H. First and final report
Lomeguatrib, a potent inhibitor of O6-alkylguanine-DNA- of a phase II study of the poly(ADP-ribose) polymerase (PARP)
alkyltransferase: phase I safety, pharmacodynamic, and inhibitor, AGO14699, in combination with temozolomide (TMZ)
pharmacokinetic trial and evaluation in combination with in patients with metastatic malignant melanoma (MM). J. Clin.
temozolomide in patients with advanced solid tumors. Clin. Cancer Pharmacol., 2006, 456.
Res., 2006, 12(5), 1577-1584. [117] Fong, P.C.; Boss, D.S.; Yap, T.A.; Tutt, A.; Wu, P.; Mergui-
[106] Ranson, M.; Hersey, P.; Thompson, D.; Beith, J.; McArthur, G.A.; Roelvink, M.; Mortimer, P.; Swaisland, H.; Lau, A.; O'Connor,
Haydon, A.; Davis, I.D.; Kefford, R.F.; Mortimer, P.; Harris, P.A.; M.J.; Ashworth, A.; Carmichael, J.; Kaye, S.B.; Schellens, J.H.; de
Baka, S.; Seebaran, A.; Sabharwal, A.; Watson, A.J.; Margison, Bono, J.S. Inhibition of poly(ADP-ribose) polymerase in tumors
G.P.; Middleton, M.R. Randomized trial of the combination of from BRCA mutation carriers. N. Engl. J. Med., 2009, 361(2), 123-
lomeguatrib and temozolomide compared with temozolomide alone 134.
in chemotherapy naive patients with metastatic cutaneous [118] Sandhu, S.K.; Yap, T.A.; de Bono, J.S. Poly(ADP-ribose)
melanoma. J. Clin. Oncol., 2007, 25(18), 2540-2545. polymerase inhibitors in cancer treatment: a clinical perspective.
[107] Watson, A.J.; Middleton, M.R.; McGown, G.; Thorncroft, M.; Eur. J. Cancer, 2010, 46(1), 9-20.
Ranson, M.; Hersey, P.; McArthur, G.; Davis, I.D.; Thomson, D.; [119] Tutt, A.; Robson, M.; Garber, J.E.; Domchek, S.M.; Audeh, M.W.;
Beith, J.; Haydon, A.; Kefford, R.; Lorigan, P.; Mortimer, P.; Weitzel, J.N.; Friedlander, M.; Arun, B.; Loman, N.; Schmutzler,
Sabharwal, A.; Hayward, O.; Margison, G.P. O(6)-methylguanine- R.K.; Wardley, A.; Mitchell, G.; Earl, H.; Wickens, M.;
DNA methyltransferase depletion and DNA damage in patients Carmichael, J. Oral poly(ADP-ribose) polymerase inhibitor
with melanoma treated with temozolomide alone or with olaparib in patients with BRCA1 or BRCA2 mutations and
lomeguatrib. Br. J. Cancer, 2009, 100(8), 1250-1256. advanced breast cancer: a proof-of-concept trial. Lancet, 2010,
[108] Watson, A.J.; Sabharwal, A.; Thorncroft, M.; McGown, G.; Kerr, 376(9737), 235-244.
R.; Bojanic, S.; Soonawalla, Z.; King, A.; Miller, A.; Waller, S.; [120] Stebbing, J.; Ellis, P.; Tutt, A. PARP inhibitors in BRCA1-
Leung, H.; Margison, G.P. Middleton, M.R. Tumor O(6)- /BRCA2-associated and triple-negative breast cancers. Future
methylguanine-DNA methyltransferase inactivation by oral Oncol., 2010, 6(4), 485-486.
lomeguatrib. Clin. Cancer Res., 2010, 16(2), 743-749. [121] Liu, X.; Han, E.K.; Anderson, M.; Shi, Y.; Semizarov, D.; Wang,
[109] Hansen, W.K.; Kelley, M.R. Review of mammalian DNA repair G.; McGonigal, T.; Roberts, L.; Lasko, L.; Palma, J.; Zhu, G.D.;
and translational implications. J. Pharmacol. Exp. Ther., 2000, Penning, T.; Rosenberg, S.; Giranda, V.L.; Luo, Y.; Leverson, J.;
295(1), 1-9. Johnson, E.F.; Shoemaker, A.R. Acquired resistance to
[110] Woolford, L.B.; Southgate, T.D.; Margison, G.P.; Milsom, M.D.; combination treatment with temozolomide and ABT-888 is
Fairbairn, L.J. The P140K mutant of human O(6)-methylguanine- mediated by both base excision repair and homologous
DNA-methyltransferase (MGMT) confers resistance in vitro and in recombination DNA repair pathways. Mol. Cancer Res., 2009,
vivo to temozolomide in combination with the novel MGMT 7(10), 1686-1692.
inactivator O(6)-(4-bromothenyl)guanine. J. Gene Med., 2006, [122] Abbotts, R.; Madhusudan, S. Human AP endonuclease 1 (APE1):
8(1), 29-34. from mechanistic insights to druggable target in cancer. Cancer
[111] Zielske, S.P.; Gerson, S.L. Lentiviral transduction of P140K Treat. Rev., 2010, 36(5), 425-435.
MGMT into human CD34(+) hematopoietic progenitors at low [123] Mendez, F.; Goldman, J.D.; Bases, R.E. Abasic sites in DNA of
multiplicity of infection confers significant resistance to BG/BCNU HeLa cells induced by lucanthone. Cancer Invest., 2002, 20(7-8),
and allows selection in vitro. Mol. Ther., 2002, 5(4), 381-387. 983-991.
[112] Tentori, L.; Leonetti, C.; Scarsella, M.; d'Amati, G.; Portarena, I.; [124] Fishel, M.L.; He, Y.; Smith, M.L.; Kelley, M.R. Manipulation of
Zupi, G.; Bonmassar, E.; Graziani, G. Combined treatment with base excision repair to sensitize ovarian cancer cells to alkylating
temozolomide and poly(ADP-ribose) polymerase inhibitor agent temozolomide. Clin. Cancer Res., 2007, 13(1), 260-267.
enhances survival of mice bearing hematologic malignancy at the [125] Liu, L.L.; Nakatsuru, Y.; Gerson, S.L. Base excision repair as a
central nervous system site. Blood. 2002, 99(6), 2241-2244. therapeutic target in colon cancer. Clin. Cancer Res., 2002, 8(9),
[113] Drew, Y.; Plummer, R. PARP inhibitors in cancer therapy: two 2985-2991.
modes of attack on the cancer cell widening the clinical [126] Taverna, P.; Liu, L.; Hwang, H.S.; Hanson, A.J.; Kinsella, T.J.;
applications. Drug Resist. Updat., 2009, 12(6), 153-156. Gerson, S.L. Methoxyamine potentiates DNA single strand breaks
[114] Plummer, E.R. Inhibition of poly(ADP-ribose) polymerase in and double strand breaks induced by temozolomide in colon cancer
cancer. Curr. Opin. Pharmacol., 2006, 6(4), 364-368. cells. Mutat. Res., 2001, 485(4), 269-281.

Received: May 25, 2010 Accepted: June 10, 2010

PMID: 22122467

You might also like