Acta Neurol Scand 2015: 132: 310–322 DOI: 10.1111/ane.12401

© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd ACTA NEUROLOGICA SCANDINAVICA

Prognostic value of epidermal growth factor receptor amplification and EGFRvIII in glioblastoma: meta-analysis Chen J-R, Xu H-Z, Yao Y, Qin Z-Y. Prognostic value of epidermal growth factor receptor amplification and EGFRvIII in glioblastoma: meta-analysis. Acta Neurol Scand 2015: 132: 310–322. © 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd. Objectives – Epidermal growth factor receptor (EGFR) gene amplification and the EGFRvIII mutation may have prognostic value in patients with glioblastoma. This meta-analysis was to determine whether EGFR gene amplification or the EGFRvIII mutation are predictors of survival in patients with glioblastoma and anaplastic astrocytoma. Materials and methods – Medline, the Cochrane Central Register of Controlled Trials, EMBASE, and Google Scholar databases were searched until July 31, 2014. Studies were selected for inclusion in the analysis if they included patients with anaplastic astrocytoma and/or glioblastoma, EGFR and/or EGFRvIII mutation status was reported, and overall survival (OS) data were reported. Results – Of 113 articles initially identified, only eight contained data with respect to the outcome of interest and were included in the metaanalysis. The number of cases ranged from 14 to 268, and the majority of patients were 60 or more years of age. There was no significant difference in OS between EGFR amplification-positive and EGFR amplification-negative glioblastoma patients (pooled hazard ratio [HR] = 1.101, 95% confidence interval [CI] 0.845, 1.434, P = 0.475) or anaplastic astrocytoma patients (pooled HR = 1.455, 95% CI 0.852, 2.482, P = 0.169). There was no significant difference in OS between EGFRvIII-positive and EGFRvIII-negative glioblastoma patients (pooled HR = 1.321, 95% CI: 0.881–1.981, P = 0.178). Significant heterogeneity existed between the studies, and the significance changed when the analysis was performed with studies removed in turn. Conclusions – There is insufficient evidence that either EGFR amplification or the EGFRvIII mutation has prognostic value in patients with glioblastoma.

Introduction

Glioblastoma multiforme (GBM) is the most common malignant primary brain tumor and has an annual incidence of approximately three cases per 100,000 persons (1, 2). Anaplastic astrocytomas (World Health Organization grade III) account for approximately 10% of all gliomas and exhibit a variable biological behavior (3, 4). Most centers believe that maximal safe resection for eligible patients followed by fractionated radiation therapy and temazolamide represent the 310

J.-R. Chen, H.-Z. Xu, Y. Yao, Z.-Y. Qin Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai, China

Key words: epidermal growth factor; receptor; epidermal growth factor receptor VIII; glioblastoma; prognosis; survival analysis Z.-Y. Qin, No 12, Middle Wulumuqi Road, Shanghai 200040, China Tel.: +86-21-38719999-2097 Fax: 86-21-50309919 e-mail: [email protected] Accepted for publication March 4, 2015

current standard of care for GBM (5, 6). The prognosis, however, is poor with an overall survival (OS) of 10–12 months (5, 7). For patients with anaplastic astrocytomas, the overall agestandardized 5- and 10-year relative survival rates are 23.6% and 15.1%, respectively (8). The most frequent genetic alteration associated with GBM is amplification of the epidermal growth factor receptor (EGFR) gene (9). Amplification results in overexpression of EGFR, a transmembrane tyrosine kinase receptor. The majority of GBMs with EGFR amplification also

EGFR and glioblastoma prognosis contain EGFRvIII, a mutant EGFR gene that is characterized by the deletion of exons 2 to 7 (9). EGFR is overexpressed in approximately 50–60% of GBMs, and EGFRvIII is found in 24–67% of cases (9, 10). Study of EGFR variations is important as tyrosine kinase inhibitors have shown promise in the treatment of GBM (11). Evidence that EGFR amplification is an independent predictor of survival in patients with GBM varies considerably between studies (1, 10, 12–18). In a previous study, EGFR amplification was not found to be a significant prognostic indicator of OS or radiographic local control in GBM patients treated with surgery (19). On the other hand, Shinojima et al. (20) reported that EGFR amplification was an independent and significant predictor of poorer OS. Other reports have indicated that EGFRvIII overexpression in the presence of EGFR amplification is the strongest indicator of poor survival (16). Contrarily, Bie nkowski et al. (14) reported that EGFRvIII expression was associated with a better prognosis. Other study has suggested that EGFRvIII status defines clinically distinct GBM subsets (21). Thus, the aim of this meta-analysis was to determine whether EGFR gene amplification or the EGFRvIII mutation is an independent predictor of survival in patients with GBM and anaplastic astrocytoma. Materials and methods Search strategy

This systematic review and meta-analysis was conducted in accordance with PRISMA guidelines (22). Medline, the Cochrane Central Register of Controlled Trials, EMBASE, and Google Scholar databases were searched until July 31, 2014 using combinations of the following terms: glioma, GBM, anaplastic astrocytoma, prognostic, prognosis, survival, EGFR, and EGFRvIII mutation. Reference lists of relevant studies were hand-searched. Selection criteria

Studies were selected for inclusion in the analysis if they included patients with anaplastic astrocytoma and/or GBM, EGFR and/or EGFRvIII mutation status was reported, and OS data were reported. Studies in which patients had primary glioma or other types of brain tumors, non-English publications, proceedings, personal communications, letters, comments,

editorials, and case reports were excluded. Studies were identified by the search strategy by two independent reviewers. When there was uncertainty regarding eligibility, a third reviewer was consulted. Data extraction

Data extraction was performed by two independent reviewers, and a third reviewer was consulted for any uncertainties. The following information was extracted from studies that met the inclusion criteria: the name of the first author, year of publication, study design, patient type/disease status, KPS, EGFR amplification and EGFRvIII mutation status, treatment, length of follow-up, and survival outcomes. Quality assessment

The modified 18-item Delphi checklist was used to assess the quality of the included studies (23). Quality assessment was performed by the independent reviewers, and a third reviewer was consulted for any uncertainties. Outcome measures and data analysis

The outcome measure was OS. Hazard ratios (HRs) with 95% confidence intervals (CIs) were calculated for OS in patients who were EGFR amplification positive or EGFRvIII positive compared to patients who were EGFR amplification negative or EGFRvIII negative, respectively. For OS, a HR > 1 indicates the HR is associated with worse survival. Heterogeneity among the studies was assessed by the Cochran Q and the I2 statistic. For Cochran Q, a value of P < 0.10 was considered to indicate statistically significant heterogeneity. For the I2 statistic, no heterogeneity was indicated when I2 = 0–25%, moderate heterogeneity when I2 = 25–50%, large heterogeneity when I2 = 50–75%, and extreme heterogeneity when I2 = 75–100%. A fixed-effects model of analysis was used for this study because of the small number of articles for each outcome (24). Sensitivity analysis using the leave-one-out approach was performed to examine the influence of individual studies on pooled estimates. Pooled HRs were calculated, and a two-sided P-value < 0.05 was considered to indicate statistical significance. All statistical analyses were performed using Comprehensive Meta-Analysis version 2.0 software (Biostat, Englewood, NJ, USA). 311

314 17.90 NA NA NA NA NA NA NA NA 6 NA NA 46 23 NA NA NA NA NA 71 NA

12.30 NA NA NA NA NA NA NA NA 10 NA NA 67 6 NA NA

NA NA NA 43 NA

+

Survival rate (%)

NA NA 19.2 6.9 22.1

13.4 (9.7, 17.0) NA 4.73 NA NA NA 504 days 12 10.5 11.1 NA NA 13.2 14.4 666 days 8

+

HR (95% CI), + vs

NA NA 33.6 34.1 22.9

11.0 (9.5, 12.6) NA 4.8 NA NA NA 458 days 12 2 9.4 NA NA 10.7 20.16 284 days 11 0.69 (0.23, 2.11) 1.24 (0.50, 3.08) 2.5 (1.1, 5.7) NA NA

NA 1.279 NA 0.92 (0.67, 1.27) 1.07 (0.17, 6.89) NA 0.69 NA NA NA 0.39 (0.46–1.40) * 0.881 NA 1.67 (1.03–2.72) NA NA NA NA NA 22 NA

8.80 NA NA NA NA 5% NA NA NA NA NA NA 59 NA NA NA

+

NA NA NA 72 NA

16.90 NA NA NA NA 21% NA NA NA NA NA NA 54 NA NA NA NA NA NA 7.2 NA

11.6 (10.6, 12.5) NA 3.27 NA NA NA NA NA NA NA NA NA 12.4 11.6 NA NA

+

NA NA NA 33 NA

11.9 (10.2, 13.6) NA 4.93 NA NA NA NA NA NA NA NA NA 12.6 16.7 NA NA

Median survival time, months (95% CI)

Median survival time, months (95% CI) Survival rate (%)

OS – EGFRvIII

OS – EGFR amplification

CI, confidence interval; HR, hazard ratio; NA, not available; OS, overall survival. Note, + and refer to EGFR amplification positive and EGFR amplification negative, or EGFRvIII positive or EGFRvIII negative. *Median (range).

Glioblastoma Weller (2014) Bienkowski (2013) Lv (2012) Srividya (2010) Viana-Pereira (2008) Pelloski (2007) Houillier (2006) Layfield (2006) Kleinschmidt-DeMasters (2005) Quan (2005) Shih (2005) Batchelor (2004) Aldape (2004) Shinojima (2003) Hurtt (1992) Torp (1992) Anaplastic astrocytoma Gulati (2010) Kouwenhoven (2009) Idbaih (2008) Aldape (2004) Smith (2001)

First author (year of publication)

Table 2 Summary of study outcomes

NA NA NA NA NA

0.75 (0.44, 1.29) 0.337 NA NA 0.73 (0.13, 4.09) 3.37 (1.74, 6.50) NA NA NA NA NA NA NA NA NA NA

HR (95% CI), + vs

Chen et al.

EGFR and glioblastoma prognosis Table 1 Summary of the studies included in the meta-analysis First author (year of publication)

Number of cases

KPS/WHO performance score

Age (years)

Male (%)

Cohort

184

Cohort Prospective phase II trial Cohort Cohort NA

83 35 140 55 268

Houillier (2006) Layfield (2006)

Cohort Retrospective

220 32

>60: 55% ≤60: 45% 60 (23, 84)* 54 (33, 73)* 47 (18, 65)* 56.1 (27, 79)† 40: 40% 40–60: 36% >60: 24%

54

62

56

KPS, Karnofsky performance status; NA, not available; RCT, randomized controlled trial; WHO, World Health Organization. *Median (range). † Mean (range).

significant different in OS between the EGFR amplification-positive and EGFR amplification-negative groups (pooled HR = 1.455, 95% CI 0.852, 2.482, Z = 1.374, P = 0.169). EGFRvIII – Three studies of glioblastoma (21, 31, 32) provided HR data for glioblastoma and EGFRvIII and were included in the analysis. As shown in Fig. 3, significant heterogeneity was present when the data from the three studies were pooled (Cochran Q = 12.47, df = 2, P = 0.002, I2 = 83.96%). The analysis revealed no significant difference in OS between EGFRvIII-positive and EGFRvIII-negative patients (pooled HR = 1.321, 95% CI: 0.881 to 1.981, Z = 1.347, P = 0.178). No studies of anaplastic astrocytoma provided sufficient EGFRvIII data for inclusion in the analysis.

Sensitivity analysis

Results of the sensitivity analysis of EGFR amplification and EGFRvIII using the leave-one-out approach are shown in Figs 4 and 5, respectively. In the analysis of glioblastoma and EGFR amplification, removal of the study by Srividya et al. (30) caused the pooled HR to become significant (P = 0.043) (Fig. 4A). With respect to anaplastic astrocytoma and EGFR amplification, the direction and magnitude of the pooled estimates did not vary considerably when the individual studies were removed in turn, indicating that the meta-analysis had good reliability (Fig. 4B). The analysis of glioblastoma and EGFRvIII showed that when the study by Weller et al. (32) was removed, the pooled HR became significant (P = 0.001; Fig. 5). 313

314 17.90 NA NA NA NA NA NA NA NA 6 NA NA 46 23 NA NA NA NA NA 71 NA

12.30 NA NA NA NA NA NA NA NA 10 NA NA 67 6 NA NA

NA NA NA 43 NA

+

Survival rate (%)

NA NA 19.2 6.9 22.1

13.4 (9.7, 17.0) NA 4.73 NA NA NA 504 days 12 10.5 11.1 NA NA 13.2 14.4 666 days 8

+

HR (95% CI), + vs

NA NA 33.6 34.1 22.9

11.0 (9.5, 12.6) NA 4.8 NA NA NA 458 days 12 2 9.4 NA NA 10.7 20.16 284 days 11 0.69 (0.23, 2.11) 1.24 (0.50, 3.08) 2.5 (1.1, 5.7) NA NA

NA 1.279 NA 0.92 (0.67, 1.27) 1.07 (0.17, 6.89) NA 0.69 NA NA NA 0.39 (0.46–1.40) * 0.881 NA 1.67 (1.03–2.72) NA NA NA NA NA 22 NA

8.80 NA NA NA NA 5% NA NA NA NA NA NA 59 NA NA NA

+

NA NA NA 72 NA

16.90 NA NA NA NA 21% NA NA NA NA NA NA 54 NA NA NA NA NA NA 7.2 NA

11.6 (10.6, 12.5) NA 3.27 NA NA NA NA NA NA NA NA NA 12.4 11.6 NA NA

+

NA NA NA 33 NA

11.9 (10.2, 13.6) NA 4.93 NA NA NA NA NA NA NA NA NA 12.6 16.7 NA NA

Median survival time, months (95% CI)

Median survival time, months (95% CI) Survival rate (%)

OS – EGFRvIII

OS – EGFR amplification

CI, confidence interval; HR, hazard ratio; NA, not available; OS, overall survival. Note, + and refer to EGFR amplification positive and EGFR amplification negative, or EGFRvIII positive or EGFRvIII negative. *Median (range).

Glioblastoma Weller (2014) Bienkowski (2013) Lv (2012) Srividya (2010) Viana-Pereira (2008) Pelloski (2007) Houillier (2006) Layfield (2006) Kleinschmidt-DeMasters (2005) Quan (2005) Shih (2005) Batchelor (2004) Aldape (2004) Shinojima (2003) Hurtt (1992) Torp (1992) Anaplastic astrocytoma Gulati (2010) Kouwenhoven (2009) Idbaih (2008) Aldape (2004) Smith (2001)

First author (year of publication)

Table 2 Summary of study outcomes

NA NA NA NA NA

0.75 (0.44, 1.29) 0.337 NA NA 0.73 (0.13, 4.09) 3.37 (1.74, 6.50) NA NA NA NA NA NA NA NA NA NA

HR (95% CI), + vs

Chen et al.

6

5

4

3

2

1

disease?

point in the

at a similar

enter the study

Did participants

consecutively?

recruited

participants

Were

appropriate?

explicit and

entry the study

criteria) to

exclusion

(inclusion and

criteria

eligibility

Are the

centre?

more than one

collected in

Were the cases

described?

study

included in the

participants

of the

characteristics

Are the

section?

methods

introduction, or

in the abstract,

clearly stated

of the study

aim/objective

hypothesis/

Is the

Y

N

Y

Y

Y

Y

N

Y

N

Y

Y

2013

2014

Y

Bienkowski

Weller

Y

N

Y

Y

Y

Y

2010

Srividya

Table 3 Modified 18-item Delphi checklist

Y

N

Y

Y

Y

Y

2009

Kouwenhoven

Y

N

Y

N

Y

Y

2008

Idbaih

Y

N

Y

N

Y

Y

2007

Pelloski

Y

N

Y

N

Y

Y

2005

Quan

Y

N

Y

Y

Y

Y

2003

Shinojima

Y

N

Y

Y

Y

Y

2001

Smith

Y

N

Y

N

Y

Y

2006

Layfield

Y

N

Y

Y

Y

Y

2012

Lv

Y

N

Y

N

Y

Y

2011

Hobbs

Y

Y

Y

N

Y

Y

2010

Gulati

Y

N

Y

N

Y

Y

2010

Coulibaly

Viana-

Y

N

Y

N

Y

Y

2008

Pereira

Y

N

Y

N

Y

Y

2006

Houillier

Y

N

Y

N

Y

Y

2005

Shih

Y

N

Y

N

Y

Y

2005

DeMasters

Kleinschmidt-

Y

Y

Y

Y

Y

Y

2004

Batchelor

Y

N

Y

N

Y

Y

1996

Waha

Y

N

Y

N

Y

Y

1995

Diedrich

Torp

Y

N

Y

N

Y

Y

1992

Y

Y

Y

Y

Y

Y

2004

Aldape

(continued)

Y

N

Y

Y

Y

Y

1992

Hurtt

EGFR and glioblastoma prognosis

315

316

13

12

11

10

9

8

7

reported?

of follow-up

Was the length

appropriate?

outcomes

the relevant

used to assess

statistical tests

Were the

intervention?

after

before and

measured

Were outcomes

methods?

or subjective

objective and/

measured with

appropriately

outcomes

Were relevant

section?

methods

introduction or

in the

clearly defined

measures

Are the outcome

in the study?

clearly reported

interventions)

(co-

interventions

Were additional

the study?

described in

clearly

intervention

Was the

Y

Y

N

Y

Y

N

Y

Y

N

Y

Y

N

Y

2013

2014

Y

Bienkowski

Weller

Table 3 (continued)

Y

Y

N

Y

Y

N

Y

2010

Srividya

Y

Y

N

Y

Y

N

Y

2009

Kouwenhoven

Y

Y

N

Y

Y

N

Y

2008

Idbaih

Y

Y

N

Y

Y

N

Y

2007

Pelloski

Y

Y

N

Y

Y

N

N

2005

Quan

Y

Y

N

Y

Y

N

Y

2003

Shinojima

Y

Y

N

Y

Y

N

N

2001

Smith

Y

Y

N

Y

Y

N

Y

2006

Layfield

Y

Y

N

Y

Y

N

Y

2012

Lv

Y

Y

N

Y

Y

N

Y

2011

Hobbs

Y

Y

N

Y

Y

N

Y

2010

Gulati

Y

Y

N

Y

Y

N

Y

2010

Coulibaly

Viana-

Y

Y

N

Y

Y

N

N

2008

Pereira

Y

Y

N

Y

Y

N

N

2006

Houillier

Y

Y

N

Y

Y

N

Y

2005

Shih

Y

Y

N

Y

Y

N

Y

2005

DeMasters

Kleinschmidt-

Y

Y

N

Y

Y

N

Y

2004

Batchelor

Y

Y

N

Y

Y

N

Y

1996

Waha

Y

Y

N

Y

Y

N

N

1995

Diedrich

Torp

Y

Y

N

Y

Y

N

Y

1992

Y

Y

N

Y

Y

N

Y

2004

Aldape

(continued)

Y

Y

N

Y

Y

N

Y

1992

Hurtt

Chen et al.

study reported?

support for the

source of

interest and

competing

Are both

results?

supported by

the study

conclusions of

Are the

reported?

events

Are adverse

outcomes?

relevant

analysis of

the data

variability in

the random

estimates of

provide

Does the study

reported?

follow-up

Was the loss to

Y, yes; N, no.

18

17

16

15

14

Y

Y

N

Y

Y

Y

N

Y

N

2013

2014

N

Bienkowski

Weller

Table 3 (continued)

Y

Y

N

Y

N

2010

Srividya

Y

Y

N

Y

N

2009

Kouwenhoven

Y

Y

N

Y

N

2008

Idbaih

Y

Y

N

Y

N

2007

Pelloski

N

Y

N

Y

N

2005

Quan

Y

Y

N

Y

N

2003

Shinojima

Y

Y

N

Y

N

2001

Smith

N

Y

N

Y

N

2006

Layfield

Y

Y

N

Y

Y

2012

Lv

Y

Y

N

Y

N

2011

Hobbs

N

Y

N

Y

N

2010

Gulati

Y

Y

N

Y

N

2010

Coulibaly

Viana-

N

Y

N

Y

Y

2008

Pereira

N

Y

N

Y

N

2006

Houillier

Y

Y

N

Y

N

2005

Shih

N

Y

N

Y

N

2005

DeMasters

Kleinschmidt-

Y

Y

N

Y

N

2004

Batchelor

N

Y

N

Y

N

1996

Waha

Y

Y

N

Y

N

1995

Diedrich

Torp

Y

Y

N

Y

N

1992

Y

Y

N

Y

N

1992

Hurtt

Y

Y

N

Y

N

2004

Aldape

EGFR and glioblastoma prognosis

317

Chen et al. A

B

Figure 2. Forest plot for the meta-analysis of overall survival of EGFR amplification for (A) glioblastoma and (B) anaplastic astrocytoma positive vs negative. CI, confidence interval.

Figure 3. Forest plot for the meta-analysis of overall survival for EGFRvIII-positive vs EGFRvIII-negative glioblastoma. CI, confidence interval.

Publication bias

As five or fewer studies are insufficient to detect funnel plot asymmetry (37), publication bias could not be assessed. Discussion

The results of this meta-analysis to determine whether presence of EGFR amplification or the EGFRvIII mutation has prognostic value in patients with GBM suggest that neither is associated with OS. However, because of the limited 318

number of studies and significant heterogeneity, the current data are not sufficient for determining whether either marker has prognostic value in patients with GBM. Despite the promise of EGFR analysis of resected tissue samples, the overwhelming effect of age, location, tumor morphology (sharp borders for infiltrative borders), resectability, and response to adjuvant treatment with radiotherapy and temazolamide are all more important with respect to prognosis. EGFR analysis may also be biased by the inherent variability in various portions of the tumor where certain cell types and genetic markers may be very heterogeneous.

EGFR and glioblastoma prognosis A

B

Figure 4. Results of sensitivity analysis using the leave-one-out approach to examine the influence of individual studies on pooled estimates of overall survival of EGFR amplification for (A) glioblastoma and (B) anaplastic astrocytoma. OR, odds ratio; CI, confidence interval.

Figure 5. Results of sensitivity analysis using the leave-one-out approach to examine the influence of individual studies on pooled estimates of overall survival for EGFRvIII-positive vs EGFRvIII-negative glioblastoma. OR, odds ratio; CI, confidence interval.

Ligand binding to the EGFR results in cell proliferation, and in normal cells, EGFR activation is tightly regulated (1). Deregulation can occur due to EGFR gene amplification or increased EGFR transcription or translation, which results in cell proliferation (1). EGFR is overexpressed in many malignancies, including those of the lung, colon, and head and neck (38). EGFRvIII is a phosphorylated EGFR mutant that results in the activation of downstream signaling pathways, and subsequent cell proliferation (9). Many studies have been performed attempting to determine whether EGFR amplification or the EGFRvIII mutation has prognostic significance in

patients with GBM, and overall, the results have been inconsistent. Early studies indicated that EGFR gene amplification was associated with a poorer prognosis in patients with GBM (33, 34). However, more recent studies and a meta-analysis have indicated that EGFR amplification had no prognostic value. Layfield et al. (16) reported that EGFR amplification status was not predictive of a favorable or unfavorable prognosis and this finding remained when the data were stratified by sex, age, and performance status. A Cleveland Clinic Foundation study also found that EGFR amplification was not a significant prognostic indicator of OS or radiographic local control in 319

Chen et al. patients with GBM (19). In contrast, Shinojima et al. (20) studied 87 patients with GBM and, on multivariate analysis, found that EGFR amplification was an independent and significant predictor of poorer OS, whereas EGFRvIII overexpression was not predictive of OS. However, in patients with EGFR amplification, EGFRvIII overexpression was an independent and significant predictor or poorer OS. Hobbs et al. (39) have suggested that the behavior of GBM and their response to therapy might vary according to the degree of amplification. A meta-analysis performed in 2000 by Huncharek and Kupelnick (18) concluded that the available data were insufficient for determining whether EGFR gene amplification is of prognostic value in GBM, and the authors pointed out that lack of control for potential confounding factors and known prognostic indicators were lacking in many studies. Younger age is considered to be associated with a better prognosis in patients with GBM. Srividya et al. (30), in a study included in the meta-analysis, found that the prognostic value of EGFR overexpression was correlated with age such that increasing age was associated with a poorer prognosis. Other studies, however, have indicated that younger patients have a poorer prognosis when the tumors exhibit EGFR amplification and that patients more than 60 years of age in which amplification is seen have a more favorable prognosis (20, 27, 39). Kleinschmidt-DeMasters et al. (17) studied 20 patients with more than 75 years of age and found that mean survival time was significantly longer in patients with EGFR amplification (8.3 months) than those without amplification (3.2 months). Disparate results have also been reported with respect to EGFRvIII expression as a prognostic indicator. Weller et al. (32) studied 184 newly diagnosed glioma patients and found that EGFRvIII status was not associated with OS or progression-free survival (PFS) and that it had no prognostic value in patients who received concomitant radiochemotherapy and were free of progression. Aldape et al. (35) examined 44 cases of GBM and found that EGFRvIII positivity was not associated with survival of GBM patients, but was highly associated with reduced survival in anaplastic astrocytoma patients. Bie nkowski et al. (14) reported that EGFRvIII expression was associated with a better prognosis (HR = 0.37) and that EGFR amplification was associated with a worse outcome in younger patients (HR = 3.75) and those that received radiotherapy (HR = 2.71). Furthermore, they 320

found that EGFR amplification was related to a better prognosis when the homozygous CDKN2A deletion was present (HR = 0.12), but to a poorer prognosis where the chromosome 7 polysomy was present (HR = 14.88). Lv et al. (15) also reported difference in survival of patients treated with cetuximab, an EGFR-blocking monoclonal antibody, who had different combinations of EGFR amplification and EGFRvIII expression. These findings underscore the complexity of the molecular pathways involved in cell proliferation, and the interactions between different mutations may help to explain disparate and sometimes contradictory results between studies. While the results of this study do not allow us to conclude whether or not the presence of EGFR amplification or the EGFRvIII mutation has prognostic value in patients with GBM, some studies have suggested that the combination of EGFR with other biomarkers or prognostic factors (e.g. age) may improve its prognostic value (36, 40). Although EGFR may not have prognostic value for GBM, other molecular markers have shown promise. Felsberg et al. (41) reported that MGMT promoter hypermethylation and near-complete tumor resection were the most important parameters associated with a better prognosis in GBM patients. A recent meta-analysis by Dong et al. (42) found that MGMT methylation was strongly correlated with longer OS and disease-free survival in patients with GBM. In another meta-analysis, Zou et al. (43) reported that isocitrate dehydrogenase isoforms 1 and 2 (IDH1/IDH2) mutations were associated with better outcomes in GBM patients. It also remains unclear whether EGFR status is valuable for guiding therapy as clinical trials have indicated that EGFR inhibitors are not effective as expected (44, 45), and investigation of the molecular characteristics of response to EGFR inhibitors is still ongoing (15, 46, 47). Wen et al. (44) studied the effect of erlotinib combined with the mechanistic target of rapamycin inhibitor temsirolimus in patients with recurrent gliomas and reported 6-month PFS rates of only 13% and 8% in GBM and AA patients, respectively. The authors postulated that the minimal antitumor activity may have been the result of insufficient tumor drug levels and a lower than expected maximum tolerated dose of the drugs as a result of increased toxicity. In a trial of bevacizumab in combination with erlotinib for recurrent gliomas, Sathornsumetee et al. (45) reported that the regimen was associated with similar radiographic response and PFS as

EGFR and glioblastoma prognosis other regimens containing bevacizumab. Lv et al. (15) found that GBM patients with EGFR amplification and without EGFRvIII expression treated with cetuximab had significantly better PFS (3.03 vs 1.63 months, P = 0.006) and OS (5.57 vs 3.97 months, P = 0.12) than those that did not receive cetuximab. Mellinghoff et al. (46) studied 49 patients with recurrent GBM and found that co-expression of EGFRvIII and phosphatase and tensin homolog (PTEN) by glioblastoma cells was associated with responsiveness to erlotinib. Similarly, Haas-Kogan et al. (47) found that GBM with high levels of EGFR expression and low levels of phosphorylated protein kinase B (PKB)/Akt had a better response to erlotinib than those with low EGFR expression and high phosphorylated PKB/Akt levels. There are limitations to this study that need to be considered. First is the small number of studies included in the analysis. While there have been many studies performed exploring the value of EGFR amplification and EGFRvIII mutation status as prognostic indicators, few of them have been of high quality and reported relevant data, as mentioned as a limitation of study in this area by the authors of the 2000 meta-analysis (20). There was significant heterogeneity among the included studies, and studies have pointed out that there is marked heterogeneity among tumors and this was not examined in the analysis. Lastly, there are different methods for determining EGFR amplification and EGFRvIII expression, and these were not taken into account in this analysis. In conclusion, the results of this meta-analysis indicate there is insufficient evidence that the presence of either EGFR amplification or the EGFRvIII mutation has prognostic value in patients with GBM. Well-designed studies that account for clinical and molecular pathological factors are necessary to determine the value of these markers in patients with GBM. Acknowledgement None.

Sources of funding None.

Conflicts of interest All authors declare no conflict of interest.

References 1. THAKKAR JP, DOLECEK TA, HORBINSKI C et al. Epidemiologic and molecular prognostic review of glioblastoma. Cancer Epidemiol Biomarkers Prev 2014;23:1985–96. 2. POSTI JP, BORI M, KAUKO T et al. Presenting symptoms of glioma in adults. Acta Neurol Scand 2015;131:88–93. 3. PALEOLOGOS NA, MERRELL RT. Anaplastic glioma. Curr Treat Options Neurol 2012;14:381–90. 4. COMPOSTELLA A, TOSONI A, BLATT V, FRANCESCHI E, BRANDES AA. Prognostic factors for anaplastic astrocytomas. J Neurooncol 2007;81:295–303. 5. STUPP R, MASON WP, VAN DEN BENT MJ et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;352:987–96. 6. JOHNSON DR, O’NEILL BP. Glioblastoma survival in the United States before and during the temozolomide era. J Neurooncol 2012;107:359–64. 7. HELSETH R, HELSETH E, JOHANNESEN TB et al. Overall survival, prognostic factors, and repeated surgery in a consecutive series of 516 patients with glioblastoma multiforme. Acta Neurol Scand 2010;122:159–67. 8. SMOLL NR, HAMILTON B. Incidence and relative survival of anaplastic astrocytomas. Neuro Oncol 2014;16:1400–7. 9. MAIRE CL, LIGON KL. Molecular pathologic diagnosis of epidermal growth factor receptor. Neuro Oncol 2014;16 (suppl 8):viii1–6. 10. HEIMBERGER AB, SUKI D, YANG D, SHI W, ALDAPE K. The natural history of EGFR and EGFRvIII in glioblastoma patients. J Transl Med 2005;3:38. 11. DE WITT HAMER PC. Small molecule kinase inhibitors in glioblastoma: a systematic review of clinical studies. Neuro Oncol 2010;12:304–16. 12. HEIMBERGER AB, HLATKY R, SUKI D et al. Prognostic effect of epidermal growth factor receptor and EGFRvIII in glioblastoma multiforme patients. Clin Cancer Res 2005;11:1462–6. 13. HOUILLIER C, LEJEUNE J, BENOUAICH-AMIEL A et al. Prognostic impact of molecular markers in a series of 220 primary glioblastomas. Cancer 2006;106:2218–23.   M, PIASKOWSKI S, STOCZYNSKA -FIDELUS E 14. BIENKOWSKI et al. Screening for EGFR amplifications with a novel method and their significance for the outcome of glioblastoma patients. PLoS One 2013;8:e65444. 15. LV S, TEUGELS E, SADONES J et al. Correlation of EGFR, IDH1 and PTEN status with the outcome of patients with recurrent glioblastoma treated in a phase II clinical trial with the EGFR-blocking monoclonal antibody cetuximab. Int J Oncol 2012;41:1029–35. 16. LAYFIELD LJ, WILLMORE C, TRIPP S, JONES C, JENSEN RL. Epidermal growth factor receptor gene amplification and protein expression in glioblastoma multiforme: prognostic significance and relationship to other prognostic factors. Appl Immunohistochem Mol Morphol 2006; 14:91–6. 17. KLEINSCHMIDT-DEMASTERS BK, LILLEHEI KO, VARELLAGARCIA M. Glioblastomas in the older old. Arch Pathol Lab Med 2005;129:624–31. 18. HUNCHAREK M, KUPELNICK B. Epidermal growth factor receptor gene amplification as a prognostic marker in glioblastoma multiforme: results of a meta-analysis. Oncol Res 2000;12:107–12. 19. QUAN AL, BARNETT GH, LEE SY et al. Epidermal growth factor receptor amplification does not have prognostic significance in patients with glioblastoma multiforme. Int J Radiat Oncol Biol Phys 2005;63:695–703.

321

Chen et al. 20. SHINOJIMA N, TADA K, SHIRAISHI S et al. Prognostic value of epidermal growth factor receptor in patients with glioblastoma multiforme. Cancer Res 2003;63:6962–70. 21. PELLOSKI CE, BALLMAN KV, FURTH AF et al. Epidermal growth factor receptor variant III status defines clinically distinct subtypes of glioblastoma. J Clin Oncol 2007;25:2288–94. 22. LIBERATI A, ALTMAN DG, TETZLAFF J et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. Ann Intern Med 2009;151:W65–94. 23. MOGA C, GUO B, SCHOPFLOCHER D et al. Development of a quality appraisal tool for case series studies using a modified Delphi technique. Edmonton, AB: Institute of Health Economics. 2012. 24. BORENSTEIN M, HEDGES LV, HIGGINS JP et al. Introduction to meta-analysis. West Sussex: Wiley, 2009;78–9. 25. KOUWENHOVEN MC, GORLIA T, KROS JM et al. Molecular analysis of anaplastic oligodendroglial tumors in a prospective randomized study: a report from EORTC study 26951. Neuro Oncol 2009;11:737–46.  E, MARIE Y et al. Gene amplification 26. IDBAIH A, CRINIERE is a poor prognostic factor in anaplastic oligodendrogliomas. Neuro Oncol 2008;10:540–7. 27. SMITH JS, TACHIBANA I, PASSE SM et al. PTEN mutation, EGFR amplification, and outcome in patients with anaplastic astrocytoma and glioblastoma multiforme. J Natl Cancer Inst 2001;93:1246–56. 28. GULATI S, YTTERHUS B, GRANLI US, GULATI M, LYDERSEN S, TORP SH. Overexpression of c-erbB2 is a negative prognostic factor in anaplastic astrocytomas. Diagn Pathol 2010;5:18. 29. SHIH HA, BETENSKY RA, DORFMAN MV, LOUIS DN, LOEFFLER JS, BATCHELOR TT. Genetic analyses for predictors of radiation response in glioblastoma. Int J Radiat Oncol Biol Phys 2005;63:704–10. 30. SRIVIDYA MR, THOTA B, ARIVAZHAGAN A et al. Age-dependent prognostic effects of EGFR/p53 alterations in glioblastoma: study on a prospective cohort of 140 uniformly treated adult patients. J Clin Pathol 2010;63:687–91. 31. VIANA-PEREIRA M, LOPES JM, LITTLE S et al. Analysis of EGFR overexpression, EGFR gene amplification and the EGFRvIII mutation in Portuguese high-grade gliomas. Anticancer Res 2008;28:913–20. 32. WELLER M, KAULICH K, HENTSCHEL B et al. Assessment and prognostic significance of the epidermal growth factor receptor vIII mutation in glioblastoma patients treated with concurrent and adjuvant temozolomide radiochemotherapy. Int J Cancer 2014;134:2437–47. 33. TORP SH, HELSETH E, DALEN A, UNSGAARD G. Relationships between Ki-67 labelling index, amplification of the epidermal growth factor receptor gene, and prognosis in human glioblastomas. Acta Neurochir (Wien) 1992;117:182–6.

322

34. HURTT MR, MOOSSY J, DONOVAN-PELUSO M, LOCKER J. Amplification of epidermal growth factor receptor gene in gliomas: histopathology and prognosis. J Neuropathol Exp Neurol 1992;51:84–90. 35. ALDAPE KD, BALLMAN K, FURTH A et al. Immunohistochemical detection of EGFRvIII in high malignancy grade astrocytomas and evaluation of prognostic significance. J Neuropathol Exp Neurol 2004;63:700–7. 36. BATCHELOR TT, BETENSKY RA, ESPOSITO JM et al. Agedependent prognostic effects of genetic alterations in glioblastoma. Clin Cancer Res 2004;10:228–33. 37. SUTTON AJ, DUVAL SJ, TWEEDIE RL, ABRAMS KR, JONES DR. Empirical assessment of effect of publication bias on meta-analyses. BMJ 2000;320:1574–7. 38. YEWALE C, BARADIA D, VHORA I, PATIL S, MISRA A. Epidermal growth factor receptor targeting in cancer: a review of trends and strategies. Biomaterials 2013;34:8690–707. 39. HOBBS J, NIKIFOROVA MN, FARDO DW et al. Paradoxical relationship between the degree of EGFR amplification and outcome in glioblastomas. Am J Surg Pathol 2012;36:1186–93.  M, BOISSELIER B, MOKHTARI K et al. Com40. LABUSSIERE bined analysis of TERT, EGFR, and IDH status defines distinct prognostic glioblastoma classes. Neurology 2014;83:1200–6. 41. FELSBERG J, RAPP M, LOESER S et al. Prognostic significance of molecular markers and extent of resection in primary glioblastoma patients. Clin Cancer Res 2009;15:6683–93. 42. DONG X, LIU RY, CHEN WD. Correlation of promoter methylation in the MGMT gene with glioma risk and prognosis: a meta-analysis. Mol Neurobiol 2014 June 10. Doi:0.1007/s12035-014-8760-3. 43. ZOU P, XU H, CHEN P et al. IDH1/IDH2 mutations define the prognosis and molecular profiles of patients with gliomas: a meta-analysis. PLoS One 2013;8:e68782. 44. WEN PY, CHANG SM, LAMBORN KR et al. Phase I/II study of erlotinib and temsirolimus for patients with recurrent malignant gliomas: North American Brain Tumor Consortium trial 04-02. Neuro Oncol 2014;16:567–78. 45. SATHORNSUMETEE S, DESJARDINS A, VREDENBURGH JJ et al. Phase II trial of bevacizumab and erlotinib in patients with recurrent malignant glioma. Neuro Oncol 2010;12:1300–10. 46. MELLINGHOFF IK, WANG MY, VIVANCO I et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med 2005;353:2012–24. 47. HAAS-KOGAN DA, PRADOS MD, TIHAN T et al. Epidermal growth factor receptor, protein kinase B/Akt, and glioma response to erlotinib. J Natl Cancer Inst 2005; 97:880–7.

Prognostic value of epidermal growth factor receptor amplification and EGFRvIII in glioblastoma: meta-analysis.

Epidermal growth factor receptor (EGFR) gene amplification and the EGFRvIII mutation may have prognostic value in patients with glioblastoma. This met...
458KB Sizes 1 Downloads 7 Views