Immunohistochemical analysis of O6-methylguanine-DNA … · 2017. 2. 8. · O6-methylguanine-DNA...

8
Full Length Article Immunohistochemical analysis of O6-methylguanine-DNA methyltransferase (MGMT) protein expression as prognostic marker in glioblastoma patients treated with radiation therapy with concomitant and adjuvant Temozolomide Samar Galal Younis a , Rasha Abd El-Ghany Khedr a, * , Safinaz Hamdy El-Shorbagy b a Clininal Oncology Department, Faculty of Medicine, Tanta University, Tanta, Egypt b Histopathology Department, Faculty of Medicine, Tanta University, Tanta, Egypt Received 26 August 2015; revised 10 November 2015; accepted 15 November 2015 Available online 10 December 2015 KEYWORDS O6-methylguanine-DNA methyltransferase; Glioblastoma; Temozolomide; Immunohistochemistry Abstract Background: O6-methylguanine-DNA methyltransferase (MGMT) protein expression using immunohistochemical analysis was proposed as a prognostic marker for patients with newly diagnosed glioblastoma (GBM) treated with radiation therapy with concurrent and adjuvant Temozolomide (TMZ). Methods: From April 2012 to October 2014, 73 patients with newly diagnosed GBM, MGMT protein expression were analyzed in formalin-fixed, paraffin-embedded tumor specimens. Patients received the radiation therapy plus concomitant and adjuvant TMZ chemotherapy. Results: For the whole cohort, the median overall survival (OS) was 15 months, and the progression-free survival was 10 months. Patients who had low MGMT protein expression (615%) had a significantly improved OS and PFS compared with patients who had high MGMT expression (17.0 months vs 14 months; P value .006) and (15.0 months vs 10 months; P value .016) respectively. The age and extent of tumor resection were the strongest clinical predictors of out- come. In multivariate Cox models MGMT protein expression, extent of tumor resection and age were identified as independent prognostic factors. * Corresponding author at: Faculty of Medicine, Tanta University, 10-Moheb Street, Mahalla Kobra, Gharbia, Egypt. Mobile: +20 01227939561. E-mail address: [email protected] (R.A.E.-G. Khedr). Peer review under responsibility of The National Cancer Institute, Cairo University. Journal of the Egyptian National Cancer Institute (2016) 28, 2330 Cairo University Journal of the Egyptian National Cancer Institute www.elsevier.com/locate/jnci www.sciencedirect.com http://dx.doi.org/10.1016/j.jnci.2015.11.003 1110-0362 Ó 2015 The Authors. Production and hosting by Elsevier B.V. on behalf of National Cancer Institute, Cairo University. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). brought to you by CORE View metadata, citation and similar papers at core.ac.uk provided by Elsevier - Publisher Connector

Transcript of Immunohistochemical analysis of O6-methylguanine-DNA … · 2017. 2. 8. · O6-methylguanine-DNA...

Page 1: Immunohistochemical analysis of O6-methylguanine-DNA … · 2017. 2. 8. · O6-methylguanine-DNA methyltransferase (MGMT) is a key enzyme in the base-excision pathway of DNA repair

Journal of the Egyptian National Cancer Institute (2016) 28, 23–30

brought to you by COREView metadata, citation and similar papers at core.ac.uk

provided by Elsevier - Publisher Connector

Cairo University

Journal of the Egyptian National Cancer Institute

www.elsevier.com/locate/jnciwww.sciencedirect.com

Full Length Article

Immunohistochemical analysis of

O6-methylguanine-DNA methyltransferase

(MGMT) protein expression as prognostic marker

in glioblastoma patients treated with radiation

therapy with concomitant and adjuvant

Temozolomide

* Corresponding author at: Faculty of Medicine, Tanta University, 10-Moheb Street, Mahalla Kobra, Gharbia, Egypt. Mobile: +20 0122

E-mail address: [email protected] (R.A.E.-G. Khedr).

Peer review under responsibility of The National Cancer Institute, Cairo University.

http://dx.doi.org/10.1016/j.jnci.2015.11.0031110-0362 � 2015 The Authors. Production and hosting by Elsevier B.V. on behalf of National Cancer Institute, Cairo University.This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Samar Galal Younis a, Rasha Abd El-Ghany Khedr a,*,

Safinaz Hamdy El-Shorbagy b

aClininal Oncology Department, Faculty of Medicine, Tanta University, Tanta, EgyptbHistopathology Department, Faculty of Medicine, Tanta University, Tanta, Egypt

Received 26 August 2015; revised 10 November 2015; accepted 15 November 2015Available online 10 December 2015

KEYWORDS

O6-methylguanine-DNA

methyltransferase;

Glioblastoma;

Temozolomide;

Immunohistochemistry

Abstract Background: O6-methylguanine-DNA methyltransferase (MGMT) protein expression

using immunohistochemical analysis was proposed as a prognostic marker for patients with newly

diagnosed glioblastoma (GBM) treated with radiation therapy with concurrent and adjuvant

Temozolomide (TMZ).

Methods: From April 2012 to October 2014, 73 patients with newly diagnosed GBM, MGMT

protein expression were analyzed in formalin-fixed, paraffin-embedded tumor specimens. Patients

received the radiation therapy plus concomitant and adjuvant TMZ chemotherapy.

Results: For the whole cohort, the median overall survival (OS) was 15 months, and the

progression-free survival was 10 months. Patients who had low MGMT protein expression

(615%) had a significantly improved OS and PFS compared with patients who had high MGMT

expression (17.0 months vs 14 months; P value .006) and (15.0 months vs 10 months; P value .016)

respectively. The age and extent of tumor resection were the strongest clinical predictors of out-

come. In multivariate Cox models MGMT protein expression, extent of tumor resection and age

were identified as independent prognostic factors.

7939561.

Page 2: Immunohistochemical analysis of O6-methylguanine-DNA … · 2017. 2. 8. · O6-methylguanine-DNA methyltransferase (MGMT) is a key enzyme in the base-excision pathway of DNA repair

24 S.G. Younis et al.

Conclusions: MGMT expression was identified as positive prognostic factor in patients with newly

diagnosed glioblastoma who underwent surgical resection followed by adjuvant radiotherapy and

concomitant oral TMZ chemotherapy (the Stupp protocol).

� 2015 The Authors. Production and hosting by Elsevier B.V. on behalf of National Cancer Institute,

Cairo University. This is an open access article under the CC BY-NC-ND license (http://creativecommons.

org/licenses/by-nc-nd/4.0/).

Aim of this work: Assessment of prognostic value ofO6-methylguanine-DNA methyltransferase (MGMT) proteinexpression using immunohistochemical analysis for patients

with newly diagnosed glioblastoma (GBM) treated with radia-tion therapy with concurrent and adjuvant Temozolomide.

Introduction

Glioblastoma (GBM) is the most common primary malignantbrain tumor in adults [1]. The worldwide incidence rate of

primary malignant brain and CNS tumors in 2012, was 3.4per 100,000. Incidence according to gender was 3.9 per100,000 in males and 3.0 per 100,000 in females. This

represented an estimated 139,608 males and 116,605 femaleswho were diagnosed worldwide with a primary malignantbrain tumor in 2012, an overall total of 256,213 individuals.The incidence rates were higher in more developed countries

(5.1 per 100,000) than in less developed countries (3.0 per100,000) [2].

In spite of multimodal intensive treatments including max-

imum surgical resection followed by radiation therapy andchemotherapy the median overall survival (MS) of patientswith GBM remains 12–15 months from the time of initial diag-

nosis [3].Temozolomide (TMZ) is an alkylating agent that is rapidly

absorbed after oral administration, and penetrates well into

the cerebrospinal fluid at a concentration up to 40% of thatmeasured in plasma [4]. TMZ has been shown to have clinicalantitumor activity against malignant gliomas and a relativelygood safety profile [5].

The European Organization for Research and Treatment ofCancer (EORTC) and the National Cancer Institute ofCanada (NCIC) conducted a trial ‘‘referred to Stupp protocol”

demonstrated that concomitant radiotherapy (RT) plus TMZfollowed by six cycles of TMZ, significantly improved the med-ian survival compared with RT alone in patients with newly

diagnosed GBM. The RT plus concomitant and adjuvantTMZ regimen has thus been considered as the standard of carefor these patients [6].

O6-methylguanine-DNA methyltransferase (MGMT) is akey enzyme in the base-excision pathway of DNA repair(BER). MGMT removes mutagenic and cytotoxic adductsfrom O6-guanine in DNA, the preferred point of attack for

alkylating drugs, such as Temozolomide (TMZ), which is usedin the treatment of glioblastoma [7].

The mechanism of action by TMZ is thought to be methy-

lation at the O6 position of guanine in DNA, with additionalmethylation at the N7 position [8,9]. These alkylating injuriesare effectively repaired by a DNA repair enzyme O6-

methylguanine-DNA methyltransferase. MGMT catalyzesthe stoichiometric, covalent transfer of the alkyl group to aninternal cysteine residue, and is finally inactivated [10].

A significant correlation was found between MGMT pro-tein expression determined by immunohistochemistry (IHC)and survival of patients with newly diagnosed GBM treated

with RT plus concomitant and adjuvant TMZ [11,12]. Themethylation of the promoter region of MGMT gene has beenshown to be a major mechanism to turn off gene transcription,

thus it may reduce the intracellular level of MGMT expression[13].

High levels of MGMT activity in tumor tissue are associ-

ated with resistance to alkylating agents [5]. In contrast, epige-netic silencing of the MGMT gene by promoter methylationresults in decreased MGMT expression in tumor cells [9,10].

At least 3 methods are available for evaluating MGMT sta-

tus: such as determining MGMT enzyme activity by usingsnap-frozen tumor samples, which may limit routine use, eval-uating MGMT protein expression by immunohistochemistry

(IHC) using paraffin-embedded tumor tissues, and establishingMGMT promoter methylation status based on methylation-specific polymerase chain reaction (MSP) or pyrosequencing

[14]. Because contradictory results have been reported acrosslaboratories based on these methods, agreement on the bestand most reliable technique for evaluating MGMT status still

needs to be achieved [15,16].

Materials and methods

Patient eligibility criteria

Between April 2012 and October 2014, 73 patients with a

newly diagnosed pathologically proven, GBM (WHO grade4 astrocytoma), in Clinical Oncology Department, TantaUniversity Hospital and Tanta Cancer Center were enrolled.

Patients were followed up until July 2015. At the time ofanalysis, the median follow up duration was 13 months(Range; 9–27 months).

Patients fulfilled the following criteria: age older than18 years, Karnofsky performance status (KPS) of P60, ade-quate bone marrow reserve (WBC count P3.5 � 109/L,

ANC countP1.5 � 109/L, platelets P100 � 109/L, and hemo-globin P9 g/dL), adequate renal function (measuredcreatinine clearance level P60 mL/min, serum creatinine61.7 mg/dl and blood urea 625 mg/dl) and adequate liver

function (transaminases less than 3 � upper normal limit,and serum bilirubin level allowed below 1.5 mg/dL).

Patients were ineligible for this study if they had metastases

to distant sites, or were pregnant or had dementia, alteredmental status, or any psychiatric condition that would prohibitthe understanding or rendering of informed consent. Also,

patients suffering from secondary malignancy or concurrentserious, uncontrolled medical illness (e.g. persistent immune-compromised states, uncontrolled infection, and clinically

significant cardiac disease) were not eligible.

Page 3: Immunohistochemical analysis of O6-methylguanine-DNA … · 2017. 2. 8. · O6-methylguanine-DNA methyltransferase (MGMT) is a key enzyme in the base-excision pathway of DNA repair

Immunohistochemical analysis of O6-methylguanine-DNA methyltransferase protein expression 25

The Ethics Committee in Faculty of Medicine, TantaUniversity, granted protocol approval and all patients signedan informed consent before the initiation of any treatment.

Study design and treatment protocol

After initial diagnosis of glioblastoma multiforme, we assigned

eligible patients for concurrent Temozolomide with radiationstarted 2–4 weeks post-surgery and paraffin blocks were col-lected for MGMT protein expression analysis, then we further

grouped patients to either low (615% positively stainednuclei) or high (>15% positively stained nuclei) proteinexpression (17–18), after concurrent chemo radiation for four

weeks patients started adjuvant Temozolomide as detailedbelow.

Surgery

All patients underwent surgery with submission of a tumor tis-sue block with a minimum of 1 cm2 of tumor for immunohisto-chemical analysis. The extent of surgical excision was defined

according to post-operative magnetic resonance imaging(MRI) or computed tomography (CT) as total resection is com-plete disappearance of contrast enhancement, subtotal resection

is disappearance of P90% of contrast enhancement, partialresection is disappearance of <90% of contrast enhancement.

Chemotherapy

Temozolomide at a dose of 75 mg/m2; 7 days per week over a40 day period with maximum 49 doses, was started along withthe radiotherapy and was continued on a daily basis until com-

pletion of radiation treatment. During the concomitant radio-therapy and Temozolomide treatment antiemetic prophylaxiswas recommended before initiation of treatment.

Adjuvant Temozolomide treatment was initiated 4 weeksafter completion of radiotherapy. Patients received Temozolo-mide at a dose of 175 mg/m2 for 5 consecutive days of a 28 day

cycle, Treatment was planned for six cycles.

Radiotherapy

Radiotherapy consisted of fractionated, conformal radiation

given at a daily dose of 2 Gy. Treatment was delivered 5 daysa week for a total of 6 weeks to a total dose of 60 Gy. Theradiotherapy protocol allowed consisted of: an initial volume

consisting of enhancement, postoperative cavity, plus sur-rounding edema (or fluid-attenuated inversion recovery[FLAIR] abnormality defined by magnetic resonance imaging

[MRI]) and a 2 cm margin received 46 Gy in 23 fractions fol-lowed by a boost of 14 Gy in seven fractions to the area ofenhancement plus the cavity and a 2.5 cm safety margin.

Paraffin blocks collection

Paraffin blocks of the eligible patients were retrieved from thearchives of Pathology Department, Faculty of Medicine,

Tanta University and Tanta Cancer Center. Hematoxylinand eosin (H and E) sections were prepared from all blocks.Informed consents for the investigational research using the

patient’s paraffin blocks were fully obtained from all patientsincluded in the study.

Immunohistochemistry

Five micrometer thick sections of 10% formalin-fixed,paraffin-embedded (FFPE) tissues were immuno-stained for

O6-methylguanine-DNA methyltransferase (MGMT). Theprimary antibody was supplied by Santa Cruz Biotechnology.Sections were deparaffinized in xylene and treated with 0.3%

hydrogen peroxide in methanol for 20 min to block endoge-nous peroxidase activity. The sections were washed in phos-phate buffered saline and then incubated with primary

antibody (dilution 1:50) over night. The slides were then incu-bated in secondary antibody. The substrate chromogen, 3.30-diaminobenzidine (DAB), enabled visualization of the nuclearexpression of MGMT via a brown precipitate. Hematoxylin

(blue) counterstaining enabled the visualization of the cellnuclei. Omission of primary antibody served as a negative con-trol. Staining of endothelial cells was used as positive internal

control.At least 100 cells were evaluated for nuclear MGMT stain-

ing. We chose areas with prominent signs of anaplasia, espe-

cially areas of high cellularity and nuclear polymorphism.Endothelial cells and perivascular lymphocytes were omittedfrom analysis.

According to Capper et al. and Lechapt-Zalcman et al. who

found that the best cut off value of MGMT protein expressionwas 15%; we divided our studied cases into low expressiongroup (615%) positively stained nuclei which was considered

immunonegative and high expression group (>15%) posi-tively stained nuclei which was considered immunopositive[17,18].

Statistical analysis

SPSS 21.0 software version (SPSS, Inc.) was used for data

analysis. To identify any selection bias when grouping patientsaccording to MGMT protein expression, baseline characteris-tics of the 2 patient groups were compared using chi-square/Fischer exact tests. Overall-survival (OS) rates were calculated

from the time of initial treatment (date of surgery) to the timeof the last follow-up visit or death. Progression-free survival(PFS) was the time elapsed from the date of initiation of treat-

ment to the date of first evidence of disease progression ordeath in the absence of disease progression. Kaplan–Meiermethod was used for estimating survival and log rank was used

to compare between the different prognostic factors. Mean andstandard deviation were used to estimate the quantitative data.Stepwise multiple regression analyses were performed using a

Cox proportional hazards model, estimating the adjusted haz-ard ratio (HR) with 95% confidence intervals (CI) for age, sex,KPS, extent of tumor resection and MGMT protein expressionrelative to the risk of death or disease progression. Statistical

tests used were two sided and the significance was consideredat values of P 6 0.05.

Results

This study was conducted between April 2012 and October2014 and recruited a total of 73 patients, including 31 females

Page 4: Immunohistochemical analysis of O6-methylguanine-DNA … · 2017. 2. 8. · O6-methylguanine-DNA methyltransferase (MGMT) is a key enzyme in the base-excision pathway of DNA repair

26 S.G. Younis et al.

and 42 males, who had a median follow-up of 13 months(range, 9–27 months).

Patient characteristics in relation to MGMT protein expres-

sion are summarized in Table 1. The mean age was 55.8 years(range, 27–76 years), and the mean KPS score was 83.4 ± SD12.5. Patients’ clinical features and tumor immunohistochemi-

cal profiles were compared (Table 1).

Progression-free survival

The median PFS was 10 months (95% CI, 7.5–12.5 months)for the whole patient series, and the PFS rate was 100%,41% and 15.4% at 6 months, 12 months and 18 months

respectively.

Figure 1 Kaplan–Meier estimates of progression-free

Table 1 Clinicopathological features of glioblastoma multi

radiation (n= 73).

Characteristics MGMT protein expression n of patients

Low expression or immunonegative 615%

n= 38 %

Age (years)

650 21 52.5

>50 17 51.5

Sex

Female 15 48.4

Male 23 54.8

KPS

P80 32 51.6

<80 6 54.5

Extent of resection

Total excision 9 50.0

Subtotal and Partial 19 52.8

Biopsy 10 52.6

n, number of patients.

KPS, Karnofsky performance score.

P value 60.05 considered significant.

In univariate analysis age, MGMT protein expression andextent of tumor excision were the only clinical factors thatinfluenced PFS: The median PFS for patients 650 years

was14 months versus 9 months for those older than 50 years(P = 0.01) whereas PFS was significantly longer for patientswith low MGMT protein expression (15 months vs 10 months)

than those with high MGMT protein expression; (P value0.016) (Fig. 1),whereas patients operated by total excisionhad longer PFS than those with biopsy only (16 m vs 8.5 m).

Other factors, such as sex and KPS did not significantly influ-ence PFS in univariate analysis (Table 2).

In the multivariate stepwise Cox proportional hazardsmodel, revealed that patients who had high MGMT protein

expression had about 2 times risk of disease progression

survival according to MGMT protein expression.

forme patients treated with Temozolomide-based chemo

73 P-value*

High expression or immunopositive >15%

n = 35 %

19 47.5 0.933

16 48.5

16 51.6 0.590

19 45.2

30 48.4 0.858

5 45.5

9 50.0 0.980

17 47.2

9 47.4

Page 5: Immunohistochemical analysis of O6-methylguanine-DNA … · 2017. 2. 8. · O6-methylguanine-DNA methyltransferase (MGMT) is a key enzyme in the base-excision pathway of DNA repair

Table 2 Association between clinical features or status of MGMT protein expression and progression free survival, assessed by

univariate analysis (log rank test).

Factors Total number Number progressed 1 year PFS (%) Median 95% CI (m) P-value

Total 73 57 41.0 10 (7.5–12.5)

Age (years) 650 40 25 56.4 14 (9.1–18.9)

>50 33 32 34.3 9 (8.6–9.4) 0.010

Sex Male 42 32 41.3 10 (8.7–11.3)

Female 31 25 40.7 11 (8–14) 0.671

KPS P80 62 48 44.8 12 (9–15)

<80 11 9 13.9 9 (8–10) 0.239

Surgery Total excision 18 13 61.1 16 (9.3–22.7)

Subtotal and partial 36 29 43.2 11 (8.6–13.4)

Biopsy 19 15 15.8 8.5 (7.9–9.1) 0.013

MGMT Ptn expression 615 38 28 59.8 15 (12–18)

>15 35 29 21.6 10 (9.4–10.6) 0.016

Median survival and 95% confidence interval, in months.

KPS, Karnofsky performance status.

Ptn, protein.

Table 3 Multivariate stepwise Cox proportional hazards

analysis for factors associated with progression.

95.0% CI for HR

HR Lower Upper P-value

Age 2.18 1.26 3.77 0.006

Surgery 0.008

Total vs subtotal 1.35 0.68 2.66 0.388

Total vs biopsy 3.33 1.48 7.45 0.004

MGMT expression 1.98 1.13 3.46 0.017

Immunohistochemical analysis of O6-methylguanine-DNA methyltransferase protein expression 27

compared with those who had low MGMT protein expression(HR, 1.98; 95% CI, 1.13–3.46; P value 0.017) (Table 3).

Overall survival

The median OS was 15 months (95% CI, 12.8–17.2 months)

for the overall study population, with OS rates of 100, 63%,32.3% and 14.8% at 6, 12, 18 and 24 months, respectively.

Figure 2 Kaplan–Meier estimates of over-all sur

In univariate analysis, the factors that influenced OS were

MGMT protein expression (17 months vs 14 months; P value.006) (Fig. 2), age at diagnosis (17 months for 650 years vs13 months for patients older than 50 years P value .014) andsurgery (18 months for total excision, 16 months for subtotal

and partial excision vs 11 month for biopsy only; P value.013) (see Fig. 3). Other factors, such as sex and KPS score,did not significantly influence OS in univariate analysis

(Table 4).In a multivariate Cox model, patients who had high

MGMT protein expression according to IHC analysis had

2.3 times risk of deaths compared with those who had lowMGMT protein expression (HR, 2.32; 95% CI, 1.3–4.15; Pvalue .004) (Table 5). At the final analysis in July 2015, 55 of

73 patients (75.3%) had died.

Discussion

GBM is an aggressive disease as no curative therapies areavailable, and few well-established prognostic factors have

vival according to MGMT protein expression.

Page 6: Immunohistochemical analysis of O6-methylguanine-DNA … · 2017. 2. 8. · O6-methylguanine-DNA methyltransferase (MGMT) is a key enzyme in the base-excision pathway of DNA repair

Figure 3 Representative photomicrographs of MGMT immunoreactivity. (A) Glioblastoma multiforme showing low MGMT nuclear

expression. (B) Another case showing low expression of MGMT, endothelial staining was used as internal positive control (arrow). (C)

Glioblastoma case with high immune-reactivity for MGMT. (D) Another case showing also high expression for MGMT. Note positive

endothelial cells as positive internal control (arrow) (� 400).

28 S.G. Younis et al.

been identified. Many recent researches in the field of neuro-oncology are directed to developing more efficient therapies,but only a small percentage of patients experience significantclinical benefit and prolonged survival [18,19].

In spite of the variability of the clinical responses, themajority of patients with GBM are presently treated in a uni-form standardized therapeutic approach, following a principle

‘one fits all’, regardless of the individual molecular characteris-tics of each tumor that most likely affect patient prognosis.Consequently, many patients display minor responses and

major therapy-related toxicities [19].

Treatment by radiotherapy plus concomitant and adjuvantTMZ for GBM has led to a small but significant improvementin patient outcome [6]; however, the responses are still verypoor and unpredictable.

In this study, our objective was to further define the prog-nostic significance of MGMT protein expression in a series ofpatients with newly diagnosed glioblastoma treated according

to the Stupp protocol (concurrent chemoradiation followed byadjuvant chemotherapy) [6,20]. MGMT expression was inves-tigated by immunostaining of paraffin sections from tumor

blocks of the studied cases.

Page 7: Immunohistochemical analysis of O6-methylguanine-DNA … · 2017. 2. 8. · O6-methylguanine-DNA methyltransferase (MGMT) is a key enzyme in the base-excision pathway of DNA repair

Table 5 Multivariate stepwise Cox proportional hazards

analysis for factors associated with survival.

95.0% CI for HR

HR Lower Upper P-value

Age 2.01 1.15 3.48 0.014

Surgery 0.005

Total vs subtotal 1.07 0.54 2.12 0.851

Total vs biopsy 2.92 1.36 6.28 0.006

MGMT expression 2.32 1.30 4.15 0.004

HR, hazard ratio.

CI, confidence interval.

Table 4 Association between clinical features or status of MGMT protein expression and overall survival, assessed by univariate

analysis (log rank test).

Survival (%)

Characteristic Total

number

Number

dead

1 year 2 years Median OS

95% CI (m)

P-value

Total 73 55 63.0 14.8 15 (12.8–17.2)

Age (years) <50 40 26 70.5 19.8 17 (14–20)

P50 33 29 79.1 9.2 13 (11–15) 0.020

Sex Male 42 31 58.3 16.9 14 (11.1–16.9)

Female 31 24 69.9 13.6 15 (11.7–18.3) 0.846

KPS P80 62 47 66.4 14.2 16 (14–18)

<80 11 8 43.6 16.4 12 (10.4–16.3) 0.258

Surgery Total excision 18 14 66.2 22.7 18 (12.3–23.7)

Subtotal and partial

excision

36 25 79.4 18.3 16 (14.5–17.5)

Biopsy 19 16 28.3 0.0 11 (10.5–11.6) 0.013

MGMT Ptn expression 615 38 26 70.2 23.4 17 (13.8–20.2)

>15 35 29 55.1 4.3 14 (10.6–17.4) 0.006

Immunohistochemical analysis of O6-methylguanine-DNA methyltransferase protein expression 29

Patient baseline characteristics were similar to thosereported in previous studies as regards age, sex, performancestatus and extent of surgery and, thus, were likely to reflectdaily practice in neuro-oncology. Age at diagnosis is a classic

prognostic factor in glioblastoma series, Our findings are inagreement with previous studies [6,17].

In current study, the extent of surgery was assessed by

postoperative imaging a significant correlation between theextent of surgery and both PFS and OS were observed inour study [6,18].

Our results indicated a median OS (15 months) for theoverall study population compared with the results reportedby Stupp et al. [21] whereas our results were less than that

reported by Lechapt-Zalcman et al. (17.5 months) because ofthe use of BCNU wafers during surgery [18]. BCNU waferswithin multimodal treatment strategies may increase survivalwith manageable adverse events attributed to slow release of

BCNU from the wafers over 2–3 weeks [18,22,23].It has been demonstrated that MGMT status is a potent

prognostic factor for patients who receive TMZ concomitant

with and adjuvant to RT and this is in agreement with thatreported by many series [17,18,24,25].

It is well known that methylation specific polymerase chain

reaction (MSP) has been proved to be a sensitive method for

assessing MGMT promoter methylation in tumor samples;which can be done on formalin-fixed, paraffin-embedded

(FFPE) tumor tissues [14]. Lechapt-Zalcman et al. also studiedMSP on FFPE glioblastoma tissue but they recommendedearly fixation in buffered formalin to prevent DNA deteriora-

tion. They added that optimal results with MSP need cryopre-served tumor specimens. However, collecting and preservingfrozen tumor specimens is costly and is not always available

in routine practice [18].Away from this argument immunohistochemistry presents

several advantages for the detection of MGMT status, pre-dominantly technical aspects (fast and easy to do, little mate-

rial required, endothelial cells used as internal positivecontrol, material can be archived in paraffin and antibodiesare commercially available). Therefore, our study, was focused

on MGMT status using immunohistochemical proteinexpression.

In our study, low protein expression (<15%) was associ-

ated independently with longer OS and PFS, suggesting thepotential usefulness of this approach in the daily evaluationof clinical samples, Different cut off values were used in previ-ous studies, ranging between 5% and 35% [17]. It is notewor-

thy that the value of 15% corresponded to the best cut offvalue for identifying better survival rates in glioblastomapatients [17,18], Capper et al. proposed that cut-off value of

15% lies beyond the observed maximum level of 7% non-neoplastic nuclear staining cells (i.e., endothelial and inflam-matory cells) within glioblastomas and they concluded that

these cells can be summed up in the group of low expressionand will not further interfere with the evaluation [17].

Sciuscio et al. performed double immunostaining with non-

tumor cell markers to differentiate glioblastoma cells fromnon-neoplastic cells. However, this procedure is costly and dif-ficult to perform as a routine pathological investigation.Another explanation for the inconsistent concordance rate

may be related to tumor heterogeneity, a particular featureof glioblastomas, which often are composed of various clonesof cells within a single tumor [16].

Our study results confirm that MGMT protein expressionis a strong and independent prognostic factor for survival in

Page 8: Immunohistochemical analysis of O6-methylguanine-DNA … · 2017. 2. 8. · O6-methylguanine-DNA methyltransferase (MGMT) is a key enzyme in the base-excision pathway of DNA repair

30 S.G. Younis et al.

patients with glioblastoma, suggesting the potential usefulnessof this approach in the daily evaluation of formalin-fixed,paraffin embedded samples.

References

[1] Preusser M, de Ribaupierre S, Wohrer A, Erridge SC, Hegi M,

Weller M, et al. Current concepts and management of

glioblastoma. Ann Neurol 2011;70:9–21.

[2] GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality

Worldwide: IARC Cancer Base No. 11 [Internet].

International Agency for Research on Cancer,

<http://globocan.iarc.fr>; 2013 [accessed 2.19.2014].

[3] Laperriere N, Zuraw L, Cairncross G. Radiotherapy for newly

diagnosed malignant glioma in adults: a systematic review.

Radiother Oncol 2002;64:259–73.

[4] Yung WK. Temozolomide in malignant gliomas. Semin Oncol

2000;27:27–34.

[5] van den Bent MJ, Taphoorn MJ, Brandes AA, Menten J, Stupp

R, Frenay M, et al. Phase II study of first-line chemotherapy

with temozolomide in recurrent oligodendroglial tumors: the

European Organization for Research and Treatment of Cancer

Brain Tumor Group Study 26971. J Clin Oncol 2003;21:2525–8.

[6] Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B,

Taphoorn MJ, et al. Radiotherapy plus concomitant and

adjuvant temozolomide for glioblastoma. N Engl J Med

2005;352:987–96.

[7] Sonoda Y, Yokosawa M, Saito R, Kanamori M, Yamashita Y,

Kumabe T, et al. O(6)-Methylguanine DNA methyltransferase

determined by promoter hypermethylation and

immunohistochemical expression is correlated with

progression-free survival in patients with glioblastoma. Int J

Clin Oncol 2010 Aug;15(4):352–8, Top of Form.

[8] Clark AS, Deans B, Stevens MF, Tisdale MJ, Wheelhouse RT,

Denny BJ, et al. Antitumor imidazotetrazines. 32. Synthesis of

novel imidazotetrazinones and related bicyclic heterocycles to

probe the mode of action of the antitumor drug temozolomide. J

Med Chem 1995;38:1493–504.

[9] Tisdale MJ. Antitumor imidazotetrazines–XV. Role of guanine

O6 alkylation in the mechanism of cytotoxicity of

imidazotetrazinones. Biochem Pharmacol 1987;36:457–62.

[10] Gerson SL. Clinical relevance of MGMT in the treatment of

cancer. J Clin Oncol 2002;20:2388–99.

[11] Chinot OL, Barrie M, Fuentes S, Eudes N, Lancelot S, Metellus

P, et al. Correlation between O6-methylguanine-DNA

methyltransferase and survival in inoperable newly diagnosed

glioblastoma patients treated with neoadjuvant temozolomide. J

Clin Oncol 2007;25:1470–5.

[12] Friedman HS, McLendon RE, Kerby T, Dugan M, Bigner SH,

Henry AJ, et al. DNA mismatch repair and O6-alkylguanine-

DNA alkyltransferase analysis and response to Temodal in

newly diagnosed malignant glioma. J Clin Oncol

1998;16:3851–7.

[13] Esteller M, Garcia-Foncillas J, Andion E, Goodman SN,

Hidalgo OF, Vanaclocha V, et al. Inactivation of the DNA-

repair gene MGMT and the clinical response of gliomas to

alkylating agents. N Engl J Med 2000;343:1350–4.

[14] Cankovic M, Mikkelsen T, Rosenblum ML, Zarbo RJ. A

simplified laboratory validated assay for MGMT promoter

hypermethylation analysis of glioma specimens from formalin-

fixed paraffin-embedded tissue. Lab Invest 2007;87:392–7.

[15] Brell M, Ibanez J, Tortosa A. O6-methylguanine-DNA methyl-

transferase protein expression by immunohistochemistry in

brain and non-brain systemic tumours: systematic review and

meta-analysis of correlation with methylation-specific

polymerase chain reaction. BMC Cancer 2011;11:35 [serial

online].

[16] Sciuscio D, Diserens AC, van Dommelen K, Martinet D, Jones

G, Janzer RC, et al. Extent and patterns of MGMT promoter

methylation in glioblastoma- and respective glioblastoma-

derived spheres. Clin Cancer Res 2011;17:255–66.

[17] Capper D, Mittelbronn M, Meyermann R, Schittenhelm J.

Pitfalls in the assessment of MGMT expression and in its

correlation with survival in diffuse astrocytomas: proposal of a

feasible immunohistochemical approach. Acta Neuropathol

2008;115:249–59.

[18] Lechapt-Zalcman E, Levallet G, Dugue AE, Vital A, Diebold

M, Menei P. O6-methylguanine-DNA methyltransferase

(MGMT) promoter methylation and low MGMT-encoded

protein expression as prognostic markers in glioblastoma

patients treated with biodegradable carmustine wafer implants

after initial surgery followed by radiotherapy with concomitant

and adjuvant temozolomide. Am Cancer Soc 2012;118:4545–54.

[19] Costa B, Caeiro C, Guimaraes I, Martinho O, Jaraquemada T,

Castro L. Prognostic value of MGMT promoter methylation in

glioblastoma patients treated with temozolomide-based

chemoradiation: a Portuguese multicenter study. Oncol Rep

2010;23:1655–62.

[20] Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N,

Weller M, et al. MGMT gene silencing and benefit from

temozolomide in glioblastoma. N Engl J Med

2005;352:997–1003.

[21] Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn

MJ, Janzer RC, et al. Effects of radiotherapy with concomitant

and adjuvant temozolomide versus radiotherapy alone on

survival in glioblastoma in a randomised phase III study: 5-

year analysis of EORTC-NCIC trial. Lancet Oncol

2009;10:459–66.

[22] Menei P, Metellus P, Parot-Schinkel E, Grogan PT, Lamont JD,

Decker PA, et al. Biodegradable carmustine wafers (Gliadel)

alone or in combination with chemoradiotherapy: the French

experience. Ann Surg Oncol 2010;17:1740–6.

[23] McGirt MJ, Than KD, Weingart JD, Chaichana KL, Attenello

FJ, Olivi A, et al. Gliadel (BCNU) wafer plus concomitant

temozolomide therapy after primary resection of glioblastoma

multiforme. J Neurosurg 2009;110:583–8.

[24] Spiegel-Kreinecker S, Pirker C, Filipits M, Lotsch D,

Buchroithner J, Pichler J, et al. O6-Methylguanine DNA

methyltransferase protein expression in tumor cells predicts

outcome of temozolomide therapy in glioblastoma patients J

Neuro Oncol 2010;12:28–36.

[25] Brell M, Ibanez J, Tortosa A. O6-methylguanine-DNA

methyltransferase protein expression by immunohistochemistry

in brain and non-brain systemic tumours: systematic review and

meta-analysis of correlation with methylation-specific

polymerase chain reaction. BMC Cancer 2011;11:35 [serial

online].