Neuro Oncology

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Neuro-Oncology 1457

23(9), 1457–1469, 2021 | doi:10.1093/neuonc/noab105 | Advance Access date 30 April 2021

MGMT promoter methylation testing to predict overall


survival in people with glioblastoma treated with
temozolomide: a comprehensive meta-analysis based
on a Cochrane Systematic Review
  

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


Sebastian Brandner , Alexandra McAleenan , Claire Kelly, Francesca Spiga , Hung-Yuan Cheng ,
Sarah Dawson, Lena Schmidt , Claire L. Faulkner, Christopher Wragg , Sarah Jefferies ,
Julian P. T. Higgins , and Kathreena M. Kurian

Division of Neuropathology and Department of Neurodegenerative Disease, UCL Queen Square Institute of
Neurology, University College London, London, UK (S.B.); Population Health Sciences, Bristol Medical School,
University of Bristol, Bristol, UK (A.M., C.K., F.S., H.Y.C., S.D., L.S., J.P.T.H.); Bristol Genetics Laboratory, Pathology
Sciences, Southmead Hospital, Bristol, UK (C.L.F., C.W.); Department of Oncology, Addenbrooke’s Hospital,
Cambridge, UK (S.F.); Bristol Medical School, Brain Tumour Research Centre, Population Health Sciences, University
of Bristol, Bristol, UK (K.M.K.)

“This article is based on a Cochrane Review published in the Cochrane Database of Systematic Reviews (CDSR): Cochrane Database
Syst Rev. 2021;3:CD013316. doi: 10.1002/14651858.CD013316.pub2.”

Corresponding Authors: Dr Kathreena M. Kurian, BSc, MD, MBBS, FRCPath(Neuro), Honorary Consultant Neuropathologist/
Associate Professor in Brain Tumour Research, Population Health Sciences, Bristol Medical School, Southmead Hospital, University
of Bristol/North Bristol Trust, Southmead Road, Bristol BS10 5NB, UK (kathreena.kurian@bristol.ac.uk); Sebastian Brandner, MD,
FRCPath, Professor of Neuropathology, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology,
Honorary Consultant Neuropathologist, The National Hospital for Neurology and Neurosurgery, University College London Hospitals
NHS Foundation Trust, Room 103 Level 1, Mailbox 126, Queen Square House, Queen Square, London WC1N 3BG, UK (s.brandner@
ucl.ac.uk).

Abstract
Background.  The DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) causes resistance of
tumor cells to alkylating agents. It is a predictive biomarker in high-grade gliomas treated with temozolomide,
however, there is no consensus on which test method, methylation sites, and cutoff values to use.
Methods.  We performed a Cochrane Review to examine studies using different techniques to measure MGMT
and predict survival in glioblastoma patients treated with temozolomide. Eligible longitudinal studies included (i)
adults with glioblastoma treated with temozolomide with or without radiotherapy, or surgery; (ii) where MGMT
status was determined in tumor tissue, and assessed by 1 or more technique; and (iii) where overall survival was
an outcome parameter, with sufficient information to estimate hazard ratios (HRs). Two or more methods were
compared in 32 independent cohorts with 3474 patients.
Results.  Methylation-specific PCR (MSP) and pyrosequencing (PSQ) techniques were more prognostic than
immunohistochemistry for MGMT protein, and PSQ is a slightly better predictor than MSP.
Conclusions.  We cannot draw strong conclusions about use of frozen tissue vs formalin-fixed paraffin-embedded
in MSP and PSQ. Also, our meta-analysis does not provide strong evidence about the best CpG sites or threshold.
MSP has been studied mainly for CpG sites 76-80 and 84-87 and PSQ at CpG sites ranging from 72 to 95. A cutoff
threshold of 9% for CpG sites 74-78 performed better than higher thresholds of 28% or 29% in 2 of the 3 good-
quality studies. About 190 studies were identified presenting HRs from survival analysis in patients in which MGMT
methylation was measured by 1 technique only.

© The Author(s) 2021. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (http://creativecommons.org/
licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
For commercial re-use, please contact journals.permissions@oup.com
1458 Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation

Key Points
• Largest meta-analysis of predictive value of MGMT test methods, cutoff
methylated/unmethylated status, and CpG sites.
• Comparison of studies using 2 or more MGMT test methods.
• Methylation-specific PCR and pyrosequencing techniques best prognosticators.

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


Importance of the Study
It is important to reach a consensus of the best promoter status assessed by a specific technique, for
method(s) for assessing MGMT methylation status, example, by pyrosequencing or methylation-specific
based on the prognostic value of each method in PCR. However, no review has quantitatively determined
predicting overall survival in people with glioblas- which method correlates best with prognosis (although
toma treated with temozolomide. Currently, there is no a previous study provided a narrative overview). In
consensus which CpG sites in the MGMT promoter our Cochrane Review, we address a research priority
region to be analyzed and which are the most rele- question identified by the James Lind Alliance Neuro-
vant cutoffs to determine methylated vs unmethylated Oncology Priority Setting Partnership—an organization
status for quantitative tests. Previous systematic re- joining academics, patients, carers, and clinicians to
views have assessed the prognostic value of MGMT set research priorities in different fields.

The IDH (isocitrate dehydrogenase) wild-type glioblastoma National Institute for Health and Care Excellence (NICE) re-
(glioblastoma multiforme [GBM]) is the most common pri- commends that all high-grade gliomas are tested.6 Most
mary brain tumor in adults, with an annual incidence of non-elderly (aged under 65  years) people are currently
approximately 3/100 000 population. The standard therapy treated with temozolomide chemotherapy regardless of
is surgical resection followed by radiotherapy and adju- MGMT promoter status, due to the lack of alternative treat-
vant treatment with temozolomide, an alkylating agent. ments.7 However, MGMT promoter status is still a useful
The median overall survival is 9.9  months for people prognostic marker that may impact clinical management
treated with surgery plus radiotherapy and 15  months for and may also be used for recruitment into clinical trials for
people treated with surgery, radiotherapy, and chemo- novel therapies.
therapy.1 For people with IDH-mutant glioblastomas, me- A number of methods have been established to as-
dian overall survival is 24  months for people treated with sess MGMT promoter methylation status: methylation-
surgery and radiotherapy, and 31 months for people treated specific PCR (MSP), quantitative (real-time) MSP, such
with surgery, radiotherapy, and chemotherapy.1 The cyto- as MethyLight MSP, pyrosequencing (PSQ), bead array,
toxic effects of temozolomide are exerted by induction of methylation-specific multiplex ligation-dependent probe
O6-methylguanine and are counteracted by the repair en- amplification (MS-MLPA)-PCR with high-resolution
zyme O6-methylguanine-DNA methyltransferase (MGMT).2 melting (HRM), co-amplification at lower denaturation
Expression of MGMT is highly regulated by epigenetic silen- temperature (COLD)-PCR, and digestion-based assays.
cing of the MGMT gene promoter and thus the MGMT pro- Immunohistochemical detection of the MGMT protein or
moter methylation status is a widely used predictive marker enzymatic activity has also been used as a proxy for meth-
for high-grade gliomas undergoing therapy with alkylating ylation status. However, internationally accepted con-
agents. However, MGMT methylation status does not al- sensus about the most appropriate diagnostic method
ways reflect gene expression, so the exact mechanism by for MGMT promoter status is lacking.8 MSP was used to
which MGMT promoter methylation improves response to assess MGMT promoter status in the landmark study by
alkylating therapy is still unknown. Hegi et al.9 The choice of technique to assess MGMT pro-
MGMT promoter methylation status testing is essen- moter status in practice also depends on the amount and
tial to inform treatment decisions in certain patients quality of the DNA sample(s) (eg, formalin-fixed paraffin-
with GBM. For example, treating elderly patients with embedded (FFPE) vs frozen tissue-derived DNA), the ro-
an unmethylated MGMT promoter with temozolomide bustness and simplicity of the method, the availability of
has been shown to be detrimental when single-agent equipment and reagents necessary for each of the tech-
temozolomide chemotherapy was compared to radio- niques, cost, and experience. In the last United Kingdom
therapy.3,4 On the basis of these findings, professional National Quality Assessment (UK NEQAS) External Quality
bodies, such as the European Association for Neuro- Assessment report, of 18 UK laboratories, 10 used PSQ,
Oncology (EANO), recommend evaluation of MGMT 5 MSP, 2 HRM, and 1 MS-MLPA. MGMT promoter meth-
promoter methylation status in elderly people,5 and The ylation can also be determined with Illumina bead chip
Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation 1459

Oncology
Neuro-
arrays, an increasingly popular method for brain tumor MS-MLPA; (vi) PCR with HRM; (vii) COLD-PCR; and (viii)
classification based on the epigenetic profile.10,11 All tech- digestion-based assays. We also included studies as-
niques can only interrogate methylation status in specific sessing (ix) MGMT expression (eg, immunohistochemistry
regions within the MGMT promoter, and the effect of meth- [IHC] for protein expression, (x) mRNA levels, or (xi) MGMT
ylation status at different sites on prognosis is not well un- enzymatic activity. Studies not reporting the test methods
derstood. In addition, some of the techniques quantify the were excluded. Studies had to report a hazard ratio (HR),
amount of methylation present, and there is no consensus or data sufficient to allow a HR to be calculated. All tech-
regarding the cutoff for categorizing methylation status. niques are listed in Table 1.
We undertook a Cochrane Review12 to assess which way
of measuring methylation of the MGMT promoter best pre-
dicts survival when people with glioblastoma are treated Search Methodology
with temozolomide. The present article provides a sum-

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


mary of the key findings from the Cochrane Review. Electronic searches were performed on the following data-
bases up to December 2018: Ovid MEDLINE, PubMed (NOT
MEDLINE), Ovid Embase, BIOSIS, and Web of Science
Conference Proceedings Citation Index (CPCI-S). No re-
Methods strictions were applied to language or date of publication.
Other resources for searches were meeting abstracts from
Study Eligibility the Society of Neuro-Oncology (SNO), EANO, and the
Japan Society for Neuro-Oncology (JSNO), retrieved via
Longitudinal studies of (i) adults (18 years and older) with the CPCI-S. We examined the reference lists of included
(ii) first occurrence or recurrent glioblastoma, (iii) treated studies and of systematic reviews that have assessed the
with temozolomide, and (iv) optionally concomitant and prognostic value of MGMT promoter status overall43 or as
adjuvant therapies in addition to temozolomide, such as assessed by a specific technique; for example, by PSQ44 or
surgery or radiotherapy or both) (v) for whom the MGMT MSP.45
status was assessed by 1 or more techniques on tumor
tissue, (vi) taken prior to treatment, but (vii) not in other
types of samples such as blood samples, or by neuro- Study Selection and Data Extraction
imaging, were eligible for inclusion. Forms of glioma other
than glioblastoma could be represented only if they consti- We used EPPI-Reviewer 4 (https://eppi.ioe.ac.uk) for pro-
tuted less than 10% of the total cases. cesses of screening and selection of studies and for part of
Eligible studies had to assess MGMT promoter meth- the data extraction the review.46 Data were extracted and
ylation status in tumor tissue by 1 or more techniques. further analyzed in Microsoft Excel. Two review authors
Eligible techniques included, but were not restricted to, (i) (“reviewers”) independently screened titles and abstracts
MSP; (ii) quantitative MSP (real-time PCR or MethyLight of all identified search results and determined whether
methylation-specific quantitative PCR); (iii) methylation- full texts should be retrieved. Then, 2 reviewers inde-
specific sequencing, including PSQ; (iv) bead array; (v) pendently assessed the full-text articles. Disagreements

  
Table 1  Summary of the Characteristics of the Included Studies Comparing 2 and More Techniques

Technique Abbreviation No. of Studies References


Pyrosequencing PSQ 20 13–27

Methylation-specific PCR MSP 17 10,13,16–19,21–37

Immunohistochemistry IHC 9 13,18–20,22,26,31,33,36,38,39

Quantitative MSP qMSP 8 13,16,19,20,24,29,37,40

PCR with high-resolution melting HRM-PCR 3 13,16,35

Bead array 2 10,41

PCR targeting mRNA PCR-mRNA 2 20,30,38

Methylation-specific multiplex ligation-dependent probe amplifica- MS-MLPA 1 34

tion
Methylation-specific restriction enzyme quantitative PCR MS-RE-qPCR 1 42

Methyl-beaming 1 42

Quantitative fluorescence immunohistochemistry QF-IHC (AQUA) 1 29

Double immunofluorescence 1 NS cohort15, RSD cohort15


qMSP combined with PSQ 1 22

qMSP combined with sequencing 1 27

Abbreviations: NS, Nordic Study; RSD, Region of Southern Denmark.


  
1460 Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation

were resolved either by consensus or by consulting a the same samples of patients. Two reviewers independ-
third reviewer. A Preferred Reporting Items for Systematic ently performed data extraction on each of these articles.
Reviews and Meta-Analyses (PRISMA) flow diagram was Disagreements were resolved by consensus, and a third
established (Figure 1) to describe the flow of information reviewer was consulted when necessary. Table 2 lists the
through the different phases of the review. items extracted.
Full data extraction, risk-of-bias assessment, and syn- We treated each method for determining MGMT pro-
thesis were performed on studies that evaluated MGMT moter methylation status as a separate prognostic factor
promoter methylation status using 2 or more methods, and extracted preferentially an unadjusted HR and its con-
enabling comparisons of methods to be made using fidence interval (CI) for each method. Where unadjusted

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


  
5494 records identified through database searching 20 additional informative
articles from references to
Ovid MEDLINE (1946 to 4-Dec-2018), n = 1500
included studies
PubMed NOT MEDLINE (4-Dec-2018), n = 101
Ovid Embase (1980 to 2018, Week 49), n = 2983
BIOSIS (1969 to 3-Dec-2018), n = 790
CPCI-S (1900 to 3-Dec-2018), n = 120

3357 record after


duplicates removed

3357 record 1590 records excluded


screened

Records excluded = 1526


Full-text articles excluded, with reasons (n = 7)
[see characteristics of excluded studies table]
Other-1 (n = 846)
[Some studies excluded with multiple reasons]
Wrong study design (67)
Wrong population (453)
MGMT measurement (241)
Wrong outcome (258)
Wrong publication type (28)
Other-2 (n = 673)
Conference abstracts (no eligible data) (608)
Single technique: HR not repoeted/can’t be
calculated (49)
Linked to another paper (5)
Other (11)
+ Awaiting Classification, n = 17
1767 full-text articles
assessed for eligibility + Ongoing Studies, n = 1

190 articles providing


223 articles includes quantitative infotmation on
in the review studies of a single method

32 seperate cohorts
(described in (33 + 20)
articles) on which multiple
methods were evaluated
using hazard ratios
(included in the
quantitative analyses)

Fig. 1  Study flow diagram illustrating the selection process of records identified in the search.
  
Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation 1461

Oncology
Neuro-
  
Table 2  Parameters Captured and Assessed for Each Included Study of 2 or More Methods

Study characteristics Author


Year
Country
Length of follow-up
Study dates
Study design
Population characteristics Number of participants

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


Population source and setting
Timing of MGMT promoter methylation assessment
Inclusion/exclusion criteria
Tumor type
Age
Gender
Karnofsky performance status
Extent of resection
Treatment regimen
Length of time between neurosurgery and start of
treatment
IDH mutation status
First diagnosis or recurrent disease
Deaths during follow-up
Prevalence of MGMT promoter methylation (by each
technique
Method(s) of MGMT promoter methylation assess- Technique
ment
Tumor sample type (ie, FFPE or frozen tissue)
Region/CpGs analyzed (for PCR-based tests); anti-
body used (for immunohistochemistry
Cutoff/threshold used to determine MGMT promoter
methylation status (where relevant)
Outcome assessment Timepoint from which overall survival is measured
Missing data Number of participants with any missing data

Abbreviations: FFPE, formalin-fixed paraffin-embedded; IDH, isocitrate dehydrogenase; MGMT, O6-methylguanine-DNA methyltransferase.
  
HRs were not reported directly, we obtained them from re- Quality in Prognosis Studies (QUIPS) tool,48 across the
ported individual participant data (IPD), reported adjusted domains: study participation, subsequent treatment, out-
HR’s or reconstructed IPD from published Kaplan-Meier come measurement, prognostic factor measurement,
survival curves.47 When IPD or reconstructed IPD available study attrition, adjustment for other potential prognostic
for 3 or more groups, the groups were combined to enable factors (where relevant), and selective reporting.
2-way comparison (eg, by comparing “unmethylated” with
combined “weakly methylated” and “methylated”). Data Synthesis and Meta-Analysis
For studies that evaluated MGMT promoter methyla-
tion status using only a single method, a single reviewer The prognostic ability of each individual method was quan-
extracted information on author, year, country, follow-up, tified using a HR for overall survival, presented with a 95%
number of participants, tumor type, IDH mutation status, CI. Comparisons of tests were restricted to those that could
and MGMT technique. be made on the same patients within the same study. The
directions of HRs were harmonized to reflect a better out-
come with a greater HR. Where 5 or more studies had com-
Assessment of Risk of Bias pared the same pair of techniques on the same patients,
we computed ratio of hazard ratios (RHR), and combined
The risk of bias in studies evaluating MGMT promoter these across studies using standard random-effects meta-
methylation status of the same patients using at least 2 analysis methods.49 We evaluated certainty in the evidence
methods was assessed with a modified version of the following the GRADE framework.50
1462 Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation

  
CpGs Technique Sym 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99
3
MS-RE-qPCR
3

1
MS-RE-qPCR
Beadarray 1
3

Almuqate 2018
1

2
MSP
Bead array
1 1 1 1 1

1
1 1 1
1

Bady 2012 (M-GBM)


Bead array
Bady 2012/Etcheverry 2010 (E-GBM)
2 2 2 2 2 2 2 2
2

2 PSQ 2 2 2 2 2

Methyl-Beaming
Barault 2015
15 15 15 15 15 15

PSQ
15
15 15 15 15 15 15 15

16 MSP 16 16 16 16 16 16 16 16 16

16

16
MSP
PSQ
16 16 16 16 16 16 16 16 16 16

Barbagallo 2014
16

14
PSQ
QF-IHC(AQUA)
14

Bell 2017
qMSP
Brigliadori 2016
14
12

12 PSQ 12 12 12 12 12 12 12 12

PSQ
Chai 2018 (7-site cohort)
12 12 12 12 12 12 12 12 12

17 PSQ 17 17 17 17

17 PSQ 17 17 17 17 17 17 17 17
17

17

17
PSQ
PSQ 17 17 17 17
17

17
17

17
17

17
17
Chai 2018 (8-site cohort)
17

17
PSQ
PSQ
17 17

17
17

17
17

17
17

17
18
Dahlrot 2018 (NS cohort)
18

18
DIF
PSQ 18 18 18 18 18
Dahlrot 2018 (RSD cohort)
DIF
Dunn 2009
18

19
18 PSQ 18 18 18 18 18

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


PSQ
Felsberg 2009
19 19 19 19 19 19 19 19 19 19 19 19 19

19 PSQ 19 19 19 19 19 19 19 19 19 19 19 19
10

19 PSQ 19 19 19 19 19 19 19 19 19 19 19 19

19

19
PSQ
PSQ
19

19
19

19
19

19
19

19
19

19
19

19
19

19
19

19
19

19
19

19
19

19
19

19
20

Havik 2012/Johannessen 2018


19

10
PSQ
IHC
19 19 19 19 19 19 19 19 19 19 19 19
21

Hsu 2015/2017
mRNA
Karayan-Tapon 2010
10

13

10 MSP
MSP
Kim 2016
20 20 20 20 20 20 20 20 20 20 20 20 20 20 20 20 20

20 MSP 20 20 20 20 20 20 20 20 20
22

20 PCR with HRM 20 20 20 20 20 20 20 20 20 20 20 20

20

20
PSQ
PSQ
20

20
20

20
20

20
20

20
20

20
9

Kristensen 2016
20

20
PSQ
PSQ
20

20
20

20
20

20
20

20
20

20
23

Lalezari 2013
Lattanzio 2015
20 PSQ 20 20 20 20 20

24

20 PSQ 20 20 20 20

PSQ
Lechapt-Zalcman2012
20 20 20 20 20

PSQ
6
20 20 20 20 20

20 PSQ 20 20 20 20

20

21
qMSP
IHC
20 20 20 20 20 20 20 20 20 20 20 20 20 20 20 4

McDonald 2013
21

21
MSP
PSQ
21

21
21

21
21

21
21

21
21 21 21 21 21
7

Melguizo 2012
qMSP
Nguyen 2015
21 21 21 21 21 21 21 21 21
8

13 IHC
MSP
Park 2011
13 13 13 13 13 13 13 13 13 13

11
13 PSQ 13

13 PSQ 13

13

13
PSQ
PSQ
13

13
26

Quillien 2012/2014 (test)


13

13
PSQ
PSQ 13 13 13 13
13

13
27

Quillien 2014 (validation)


qRT-PCR
Quillien 2016/2017
13
25

13 SQ-MSP 13 13 13 13 13 13 13 13 13

MSP
Thon 2017
22 22 22 22 22 22 22 22 22 22

29
22 PSQ 22 22 22 22 22 22 22

IHC
Yamashita 2018
9

9 PSQ 30

9 qMSP-PSQ
9

9
qMSP-PSQ
qMSP-PSQ
31

Yang 2012
23

23
IHC
MSP 23 23 23 23 23 23 23 23 23
32

Yoshioka 2018
23 PSQ 23 23 23 23 23 23 23 23 23 23 23 23 23 23 23 23 23 23 23 23 23 23 23 23

MSP

Test Symbol
24 24 24 24 24 24 24 24 24 24

24 MSP 24 24 24 24 24 24 24 24 24

24 PSQ 24 24 24 24 24 24 24 24 24

24 PSQ 24 24 24 24 24 24 24 24 24

6
IHC
MSP 6 6 6 6 6 6 6 6
Beadarray
4 MSP 4 4 4 4 4

7
PSQ
IHC
4 4 4 4 4

DIF
7 MSP 7 7 7 7 7 7 7 7

8
FSQ-MS-PCR
FSQ-MS-PCR
8

8
8

8
8

8
8

8
8

8
8

8
8

8
8

8
8

8
FSQ-MS-PCR
11 MS-MLPA
11

11
MS-MLPA
MSP 11 11 11 11 11 11 11 11
IHC
25 PSQ 25 25 25 25 25

25

25
PSQ
PSQ
25

25
25

25
25

25
25

25
25

25
MethyI-Beaming
25 PSQ 25 25 25 25 25

25

25
PSQ
PSQ
25

25
25

25
25

25
25

25
25

25
MethylLight-MSP
25 PSQ 25 25 25 25 25

25

25
PSQ
PSQ
25

25
25

25
25

25
25

25
25

25
mRNA
25 PSQ 25 25 25 25 25

25

25
PSQ
PSQ
25

25
25

25
25

25
25

25
MS-MLPA
25
SQ-MSP 25 25 25 25 25 25 25 25 25

SQ-MSP
MSP
25 25 25 25 25 25 25 25 25 25

25
SQ-MSP 25 25 25 25 25 25 25 25 25

25
SQ-MSP 25 25 25 25 25 25 25 25 25

MS-RE-qPCR
25 SQ-MSP 25 25 25 25 25 25 25 25 25

26 IHC
26
MethylLight-MSP 26 26 26 26 26 26 26 26 26 26 26 26

PCR with HRM


26 MSP 26 26 26 26 26 26 26 26 26

26 PCR with HRM 26 26 26 26 26 26 26 26 26 26 26 26 26 26

26 PSQ 26

PSQ
26 PSQ 26

26 PSQ 26

26 PSQ 26

PSQ
QF-IHC(AQUA)
26 26

26 PSQ 26

26 PSQ 26

PSQ
qMSP
26 26

26 PSQ 26

26 PSQ 26

PSQ
qMSP-PSQ
26 26

26 PSQ 26

26 PSQ 26

PSQ
qRT-PCR
26 26

26 PSQ 26

26 PSQ 26

PSQ
SQ-MSP
26 26

26 PSQ 26

26 PSQ 26

26 PSQ 26

26 PSQ 26 26 26 26 26

26 PSQ 26 26 26 26 26

26 PSQ 26 26 26 26 26 26 26 26 26 26 26 26 26 26 26 26

26 PSQ 26 26 26 26 26

26 PSQ 26 26 26 26

26 PSQ 26 26 26 26 26

26 PSQ 26 26 26 26 26

26 PSQ 26 26 26 26 26

26 PSQ 26 26 26 26 26

26 PSQ 26 26 26 26 26

26 PSQ 26 26 26 26 26 26

26 PSQ 26 26 26 26 26

27 PSQ 27 27 27 27 27

27 PSQ 27 27 27 27 27

27 PSQ 27 27 27 27 27

29 MSP 29 29 29 29 29 29 29 29 29

29 PSQ 29 29 29 29 29 29 29 29 29 29 29 29 29 29 29 29 29 29 29 29 29 29 29 29 29

30 MSP 30 30 30 30 30 30 30 30 30

30 PCR with HRM 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30

30 PCR with HRM 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30

30 PCR with HRM 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30

30 PCR with HRM 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30

30 PCR with HRM 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30 30

31 IHC
31 MSP 31 31 31 31 31 31 31 31 31

32 MSP 32 32 32 32 32 32 32 32 32

32 MSP 32 32 32 32 32 32 32 32 32

32 MSP 32 32 32 32 32 32 32 32 32

32 MSP 32 32 32 32 32 32 32 32 32

32 MSP 32 32 32 32 32 32 32 32 32

Fig. 2  Schematic overview of the CpG sites tested in the different publications. The first column is a color-coded representation of the authors,
which are shown in the inset on the right. The CpG sites are listed in numerical order, corresponding to the iterative positions relative to transcrip-
tion start. The corresponding sites, test methods, and thresholds are shown in detail in the Supplementary data. Each row represents a distinct
method and where applicable, different CpG sites or thresholds. Rows with blank cells (ie, no color-coded CpG sites) indicate that a method was
not PCR-based test or that CpG information is not available. For studies using PCR primers as described by Esteller et al.51 CpG site location is
based on Malley et al.52
  
Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation 1463

Oncology
Neuro-
Additional Analyses America (n  =  2), East Asia (n  =  8), Australia (n  =  1), or in
multiple countries (n = 2). Average patient age ranged from
The full Cochrane Review includes more details of the 44 to 64, with an overall male: female ratio of 1.5:1. The vast
methods and further analyses including adjusted HRs majority were patients with glioblastomas, predominantly
(examining the prognostic value of tests in addition to age undergoing total resection. Figure 2 illustrates the CpG
and extent of resection) and sensitivity analyses. In addi- sites targeted in the studies. The Supplementary data pro-
tion, it collates information about the UK costs of the main vide a comprehensive overview of the data from all indi-
techniques and cost comparison ratios.12 vidual comparison studies.

Findings: Comparisons of Different Techniques


Results

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


The 160 extracted HRs are reported in the Supplementary
Results of the Search data and summarized in Table 3. In all cases, the estimated
HR is above 1, indicating higher hazard of death in those
The search identified 5494 records, of which 223 were in- with unmethylated MGMT promoters. In the vast majority
cluded in the review (see Figure 1). These comprised 32 of cases, the lower limit of a 95% CI for the HR is above 1,
separate cohorts of patients (“studies”) where 2 or more confirming the prognostic value of MGMT promoter meth-
methods were compared, including studies comparing ylation status. When examining these results, we empha-
different variants of the same technique. About 190 fur- size that comparisons should only be made of different
ther articles describing single-technique studies were methods within studies. HRs should not be compared
also included and are described in a separate section across studies because there are many (more substantial)
below. differences between these results than the choice of tech-
nique, tumor sample, CpG islands, or thresholds.
Characteristics of the Included Studies Meta-analysis of RHR (Table 3) shows that MSP (CpG
sites 76-80 and 84-87) is more prognostic than IHC (varying
The 32 studies included a total of 3474 participants. The thresholds) with RHR  =  1.31 (95% CI: 1.01-1.71). Since a
techniques investigated and the corresponding references large majority of MSP studies had examined CpG sites
are listed in Table 1. All studies had a standard cohort de- 76-80 and 84-87,52 we were unable to compare alternative
sign. Studies were undertaken in Europe (n  =  19), North CpG sites for MSP. We also found evidence that PSQ is

  
Table 3  Summary of Findings of Comparisons of Methods for Measuring MGMT Promoter Methylation Status

Technique 1 Technique 2 RHR (95% CI) Partici- Studies Certainty of Reason for Down Rating
pants Evidence
MSP IHC 1.31 (1.01-1.71) 913 7 Moderate Imprecision
PSQ IHC 1.36 (1.01-1.84) 871 5 Low Imprecision and indirectness (due to variability in
CpG sites and thresholds used for PSQ)
PSQ MSP 1.14 (0.87-1.48) 1119 9 Low Imprecision and indirectness (due to variability in
CpG sites and thresholds used for PSQ)
PSQ PSQ (variant Not estimated 876 11 Very low Serious risk of bias, imprecision, inconsistency,
of) and indirectness
qMSP MSP of PSQ Not estimated 765 7 Very low Risk of bias, imprecision, inconsistency, and indi-
rectness
Bead array MSP of PSQ Not estimated 81 2 Very low Serious imprecision, inconsistency, and indirect-
ness
PCR-mRNA MSP or PSQ Not estimated 148 2 Very low Imprecision, inconsistency, and indirectness
MS-MLPA MSP or PSQ Not estimated 48 1 Very low Serious risk of bias, serious imprecision, incon-
sistency, and indirectness
PCR-HRM MSP or PSQ Not estimated 309 3 Very low Risk of bias, serious imprecision, inconsistency,
and indirectness
Others MSP or PSQ Not estimated 1209 7 Very low Serious imprecision, inconsistency, and indirect-
ness

Abbreviations: CI, confidence interval; RHR, ratio of hazard ratios; for technique abbreviations, see Table 2.
The outcome being predicted is overall mortality (time to death). Grades of evidence: high quality, further research is very unlikely to change our con-
fidence in the conclusion; moderate quality, further research is likely to have an important impact on our confidence in the conclusion; low quality,
further research is very likely to have an important impact on our confidence in the conclusion; very low quality, we are very uncertain about the
conclusion.
  
1464 Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation

more prognostic than IHC (RHR = 1.36; 95% CI: 1.01-1.84), (RHR = 1.14; 95% CI: 0.87-1.48). The CpG sites targeted by
although studies of PSQ feeding into this analysis had tar- PSQ ranged between 72 and 95, and several studies had
geted different CpG sites. While there is a consistent pat- examined sites 74-78. There was a suggestion that PSQ
tern that PSQ seems to be a slightly better predictor than (mainly at CpG sites 74-78, but with varying thresholds) is
MSP, there is no strong statistical evidence to confirm this slightly more prognostic than MSP (sites 76-80 and 84-87).

  
Risk of bias
Colour code Study ID (author/year) Participant Subsequent Outcome
selection treatment measurement

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


Almuqate 2018

Bady 2012 (M-GBM)

Bady 2012/Etcheverry 2010 (E-GBM)

Barault 2015

Barbagallo 2014

Bell 2017

Brigliadori 2016

Chai 2018 (7-site cohort)

Chai 2018 (8-site cohort)

Dahlrot 2018 (NS cohort)

Dahlrot 2018 (RSD cohort)

Dunn 2009

Felsberg 2009

Havik 2012/Johannessen 2018

Hsu 2015/2017

Karayan-Tapon 2010

Kim 2016

Kristensen 2016

Lalezari 2013

Lattanzio 2015

Lechapt-Zalcman 2012

McDonald 2013

Melguizo 2012

Nguyen 2015

Park 2011

Quillien 2012/2014 (test)

Quillien 2014 (validation)

Quillien 2016/2017

Thon 2017

Yamashita 2018

Yang 2012

Yoshioka 2018

Fig. 3  Study-level risk-of-bias assessments for studies comparing 2 or more methods. participant selection, subsequent treatment, and out-
come. Green (+) = low risk of bias; Yellow (−) = unclear risk of bias. The color codes of the individual studies correspond to those in Figure 1.
Abbreviations: GBM, glioblastoma multiforme; NS, Nordic Study; RSD, Region of Southern Denmark.
  
Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation 1465

Oncology
Neuro-
We did not perform formal analyses to investigate Risk-of-Bias Assessment and Certainty in the
whether heterogeneity in HRs may have been due to age, Evidence
extent of tumor resection, Karnofsky performance status,
IDH status, first diagnosis vs recurrence, start and length We present results of the risk-of-bias assessment for the
of follow-up, due to the very limited replication of specific 3 domains that apply to the whole studies in Figure 3. All
methods, and large amounts of missing data for many of studies were assessed to be at low or unclear risk of bias for
these study characteristics. participant selection. All studies except one were assessed
Many variants of PSQ have been compared, although we as at low risk of bias arising due to variation in subsequent
did not see any strong and consistent messages from the treatment after collection of the tumor sample. All studies
results. Thresholds varied substantially (from 4% to 25% were assessed to be at low risk of bias in measurement of
for single CpG sites; and from 2.68% to 35% for multiple the outcome (all-cause mortality). The other aspects of the
CpG sites). Two of the three studies with low (or unclear) risk-of-bias assessment apply to individual results. We were

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


risk of bias that compared different thresholds found that a mostly free of concerns about risk of bias in the domains
9% threshold was more prognostic than higher thresholds for study attrition, problems with other prognostic factors
(of 28% or 29%; see top 2 results in Figure 4). We are un- adjusted for, and selective reporting. For some results, the
able to draw strong conclusions about use of frozen tissue threshold used to classify methylation status was derived
vs FFPE in MSP, although 1 study observed that MSP was from the data, leading to a high risk of bias. The result-level
more prognostic when based on frozen tissue. No clear risk-of-bias assessments for studies examining PSQ are in-
difference was apparent between using PSQ on FFPE vs cluded in Figure 4. Table 3 summarizes the certainty of the
frozen tissue. evidence from comparative studies, grouped by technique.

  
Table 4  Characteristics of Studies Examining MGMT Promoter Methylation With 1 Technique Only

Study Parameter Characteristics No. of Studies


Total number of studies 190
Reporting follow-up information 54
Reporting follow-up range 29
Reporting data on IDH1/IDH2 mutation 62
All IDH wild type 11
IDH mutation present (0.7%-73.4%) 47
No IDH mutation reported 3
Reporting tumor type Glioblastomas only (all studies) 183
Glioblastoma: supratentorial 9
Glioblastoma: primary 23
Glioblastoma: primary, supratentorial 1
Glioblastoma: recurrent 4
Mixed glioma + gliosarcoma 6
Gliosarcoma only 1
Test method MSP 94
PSQ 27
qMSP (real-time PCR or MethyLight) 22
Bead array 10
MS-MLPA 4
HRM-PCR 3
MGMT protein (IHC) 21
MGMT protein (Western blot) 1
mRNA 4

Abbreviations: HRM-PCR, PCR with high-resolution melting; IDH, isocitrate dehydrogenase; IHC, immunohistochemistry; MGMT, O6-
methylguanine-DNA methyltransferase; MS-MLPA, methylation-specific multiplex ligation-dependent probe amplification; MSP, methylation-specific
PCR; PSQ, pyrosequencing; qMSP, quantitative methylation-specific PCR.
As per the study protocol, the results of these studies were not examined, because comparisons of HRs across studies would not provide reliable
indicators of differences between the methods.
  
1466 Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation

  
Study ID and CpGsanalysed Threshold for Hazard ratio PF Selective
Sample type (PCR-based tests) methylated (95% CI) measurement Attrition reporting
Barbagallo 2014
FFPE NR >9% 3.73 (1.68, 8.28) Low RoB Low RoB Low RoB
FFPE NR >25% 1.99 (0.92, 4.32) Low RoB Low RoB Low RoB

Brigliadori 2016
FFPE 74-83 >9% 1.92 (1.17, 3.14) Low RoB Low RoB Low RoB
FFPE 74-83 >29% 3.02 (1.72, 5.29) Low RoB Low RoB Low RoB

Chai 2018 (7-site cohort)


Frozen 72-78 >12% 2.94 (1.12, 7.69) High RoB Low RoB Unclear
Frozen 74-78 >12% 2.94 (1.12, 7.69) High RoB Low RoB Unclear
Frozen 75-78 >12% 2.94 (1.12, 7.69) High RoB Low RoB Unclear

Chai 2018 (8-site cohort)


Frozen 75-78 >13% 2.70 (1.37, 5.26) High RoB Low RoB Unclear
Frozen 75-82 >12% 3.03 (1.54, 6.25) High RoB Low RoB Unclear
Frozen 76-79 >11% 2.13 (1.09, 4.17) High RoB Low RoB Unclear

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


Dunn 2009
Frozen, smear and/or FFPE 72-83 >9% 3.57 (2.24, 5.70) Low RoB Low RoB Low RoB
Frozen, smear and/or FFPE 72-83 >20% 4.25 (2.57, 7.05) Unclear Low RoB Low RoB
Frozen, smear and/or FFPE 72-83 >29% 4.03 (2.30, 7.07) High RoB Low RoB Low RoB
Frozen, smear and/or FFPE 72-83 >35% 3.64 (1.99, 6.67) Unclear Low RoB Low RoB
Frozen, smear and/or FFPE 72-83 Cluster 1 vs 2&3 4.44 (2.58, 7.66) Unclear Low RoB Low RoB
Frozen, smear and/or FFPE 72-83 Cluster 1&2 vs 3 3.59 (2.26, 5.69) Unclear Low RoB Low RoB

Havik 2012
Frozen 74-78 >2.68% 1.85 (1.02, 3.35) Low RoB Low RoB Low RoB
Frozen 74-78 >6% 2.30 (1.21, 4.38) High RoB Low RoB Low RoB
Frozen 74-78 >7% 2.33 (1.19, 4.57) High RoB Low RoB Low RoB
Frozen 74-78 >8% 2.33 (1.19, 4.57) High RoB Low RoB Low RoB
Frozen 74-78 >9% 2.30 (0.99, 5.33) High RoB Low RoB Low RoB
Frozen 76-79 >6% 2.30 (1.21, 4.38) High RoB Low RoB Low RoB
Frozen 76-79 >7% 2.33 (1.19, 4.57) High RoB Low RoB Low RoB
Frozen 76-79 >8% 1.90 (0.99, 3.65) High RoB Low RoB Low RoB
Frozen 76-79 >9% 1.90 (0.99, 3.65) High RoB Low RoB Low RoB

Karayan-Tapon 2010
Frozen 74 >5.5% 3.26 (1.91, 5.59) Low RoB Low RoB Low RoB
Frozen 74-78 >8.0% 3.35 (1.95, 5.73) Low RoB Low RoB Low RoB
Frozen 75 >8.7% 3.26 (1.91, 5.57) Low RoB Low RoB Low RoB
Frozen 76 >8.0% 2.78 (1.64, 4.74) Low RoB Low RoB Low RoB
Frozen 77 >7.85% 3.65 (2.10, 6.34) Low RoB Low RoB Low RoB
Frozen 78 >7.8% 2.70 (1.59, 4.58) Low RoB Low RoB Low RoB

Lattanzio 2015
FFPE 72-80 ?9% 2.09 (1.09, 3.99) Low RoB Low RoB Low RoB
Frozen 72-80 ?9% 2.25 (1.19, 4.25) Low RoB Low RoB Low RoB

Quillien 2014 (test)


Frozen 74 >4% 2.44 (1.51, 3.95) High RoB Low RoB Low RoB
Frozen 74 >8% 2.22 (1.30, 3.79) High RoB Low RoB Low RoB
Frozen 74-78 >8% 3.13 (1.86, 5.25) High RoB Low RoB Low RoB
Frozen 74-78 >9% 3.13 (1.81, 5.38) High RoB Low RoB Low RoB
Frozen 74-89 >11% 3.13 (1.84, 5.30) High RoB Low RoB Low RoB
Frozen 75 >11% 3.23 (1.87, 5.55) High RoB Low RoB Low RoB
Frozen 75-79 >8% 2.94 (1.71, 5.06) High RoB Low RoB Low RoB
Frozen 76 >4% 2.63 (1.61, 4.31) High RoB Low RoB Low RoB
Frozen 76 >5% 2.78 (1.62, 4.75) High RoB Low RoB Low RoB
Frozen 76-79 >8% 2.86 (1.66, 4.92) High RoB Low RoB Low RoB
Frozen 76-80 >9% 3.03 (1.74, 5.26) High RoB Low RoB Low RoB
Frozen 77 >6% 3.13 (1.87, 5.23) High RoB Low RoB Low RoB
Frozen 77 >7% 2.78 (1.63, 4.74) High RoB Low RoB Low RoB
Frozen 77-81 >8% 2.70 (1.59, 4.59) High RoB Low RoB Low RoB
Frozen 78 >4% 2.33 (1.38, 3.92) High RoB Low RoB Low RoB
Frozen 78 >5% 2.50 (1.51, 4.13) High RoB Low RoB Low RoB
Frozen 78-82 >9% 2.86 (1.68, 4.86) High RoB Low RoB Low RoB
Frozen 79 >7% 2.78 (1.61, 4.80) High RoB Low RoB Low RoB
Frozen 79-83 >8% 2.86 (1.68, 4.86) High RoB Low RoB Low RoB
Frozen 80 >4% 2.56 (1.54, 4.28) High RoB Low RoB Low RoB
Frozen 81 >8% 2.44 (1.44, 4.14) High RoB Low RoB Low RoB
Frozen 82 >16% 2.94 (1.70, 5.08) High RoB Low RoB Low RoB
Frozen 83 >10% 2.78 (1.65, 4.66) High RoB Low RoB Low RoB
Frozen 84 >9% 3.23 (1.86, 5.59) High RoB Low RoB Low RoB
Frozen 84-88 >17% 3.23 (1.86, 5.58) High RoB Low RoB Low RoB
Frozen 84-89 >22% 3.23 (1.85, 5.62) High RoB Low RoB Low RoB
Frozen 85 >5% 2.50 (1.52, 4.11) High RoB Low RoB Low RoB
Frozen 85-89 >13% 2.94 (1.76, 4.92) High RoB Low RoB Low RoB
Frozen 86 >11% 2.78 (1.65, 4.69) High RoB Low RoB Low RoB
Frozen 87 >25% 3.03 (1.75, 5.24) High RoB Low RoB Low RoB
Frozen 88 >4% 2.27 (1.38, 3.75) High RoB Low RoB Low RoB
Frozen 89 >12% 3.23 (1.91, 5.46) High RoB Low RoB Low RoB

Quillien 2014 (validation)


FFPE 74-78 >9% 3.70 (1.71, 8.01) Low RoB Low RoB Low RoB
FFPE 74-78 >10% 2.86 (1.42, 5.74) Low RoB Low RoB Low RoB
FFPE 74-78 >28% 2.27 (0.98, 5.29) Low RoB Low RoB Low RoB

Quillien 2016
FFPE 74-78 >6% 3.23 (2.02, 5.16) High RoB Unclear Low RoB
FFPE 74-78 >8% 4.00 (2.30, 6.97) Low RoB Unclear Low RoB
FFPE 74-78 >12% 4.17 (2.35, 7.38) Unclear Unclear Low RoB
FFPE 74-78 >13% 4.35 (2.41, 7.83) Unclear Unclear Low RoB
FFPE 74-78 >16% 4.55 (2.48, 8.34) High RoB Unclear Low RoB
Frozen 74-78 >6% 4.00 (2.30, 6.97) High RoB Unclear Low RoB
Frozen 74-78 >8% 3.57 (2.14, 5.95) Low RoB Unclear Low RoB
Frozen 74-78 >12 or 13% 3.45 (2.10, 5.66) Unclear Unclear Low RoB
Frozen 74-78 >12% 3.70 (2.19, 6.26) Low RoB Unclear Low RoB
Frozen 74-78 >16% 3.13 (1.94, 5.04) High RoB Unclear Low RoB
Frozen 76-79 >8% 3.33 (2.06, 5.40) Low RoB Unclear Low RoB
Frozen 76-79 >12% 3.33 (2.06, 5.40) Low RoB Unclear Low RoB

.5 1 2 4

Fig. 4  Hazard ratios from studies comparing different methods for PSQ. Hazard ratios from studies comparing different methods for PSQ. The
scale on the bottom of the figure indicates the hazard ratio. Abbreviations: CI, confidence interval; CpG, 5′-cytosine-phosphate-guanine-3′; FFPE,
formalin-fixed paraffin-embedded; NR, not reported; PF, prognostic factor; PSQ, pyrosequencing; RoB, risk of bias.
  
Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation 1467

Oncology
Neuro-
Studies Examining Only a Single Technique varied markedly across studies, and we were unwilling to
make naive indirect comparisons of techniques across dif-
About 190 articles described studies presenting HRs from ferent studies and we are presenting quantitative results
survival analysis in patients in which MGMT methylation for these studies.12
was measured by 1 technique, and studies in which more We rated the evidence for the comparison between MSP
than 1 technique was used but only MGMT methylation data and IHC as of “moderate certainty,” and the evidence for
from 1 method were used in the survival analysis (Table 4). comparisons of PSQ with MSP or IHC as of “low certainty”
These studies included a total of 27 710 participants (range (Table 3). All other comparisons we rated as “very low cer-
6-1395). They were conducted in Italy (n = 29), multiple coun- tainty.” Although risk-of-bias and publication bias were not
tries (n = 23), Germany (n = 21), the United States (n = 20), major concerns for us, we rated down many of our assess-
Japan (n = 18), China (n = 17), South Korea (n = 11), France ments because there was a wide variety of different CpG
(n = 9), Denmark (n = 8), Spain (n = 8), the United Kingdom sites and thresholds investigated, without systematic repli-

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


(n = 6), India (n = 3), Switzerland (n = 3), Australia, Belgium, cations of findings using the same methods across studies.
Czech Republic, Egypt, Taiwan (n = 2), and 1 study each in The amount of evidence is small, with only tens or at most
Canada, Portugal, Netherlands, and Tunisia. the low hundreds of participants contributing to evidence
for many of the techniques.
The evidence identified was generally applicable to clinical
Discussion practice. We included only studies in which at least 90% of pa-
tients had glioblastoma, and nearly all patients were treated
We took a systematic approach to identifying, appraising, with temozolomide. We focused on overall survival only, so
and collecting information from the evidence and assessed are unable to draw conclusions about using techniques to
risk of bias and applicability concerns using a modification predict progression-free survival. The decision which method
of QUIPS specific to the topic of the Cochrane Review.48 to use in clinical practice however is not necessarily guided
This is the first systematic review to our knowledge that by best predictive value but is influenced by cost, turnaround
compares methods for categorizing tumors as methylated time, availability of equipment: PSQ, the most quantitative
in relation to their ability to predict survival in patients method can be limited by the availability of equipment, while
with glioblastoma. Unsurprisingly, among methods for qMSP, a commonly used method, cannot accurately quantify
assessing MGMT status in glioblastoma patients treated heterogeneously methylated CpG sites.
with temozolomide, PSQ and MSP appear to be more Further large studies examining the use of different tech-
prognostic for overall survival than IHC. While there is a niques, using pre-defined threshold values for interpre-
consistent pattern that PSQ seems to be a slightly better tation, would provide valuable new information on these
predictor than MSP, there is no strong statistical evidence methods, and our review reflects the reality that it may be
to confirm this. Moreover, there is no strong evidence to challenging to reach a consensus for the best method of
draw conclusions with confidence about the best CpG sites MGMT promoter methylation testing.
or thresholds for quantitative methods. In our study, MSP
has been studied mainly for CpG sites 76-80 and 84-87 and
PSQ at CpG sites ranging from 72 to 95. A cutoff threshold
of 9% for CpG sites 74-78 was found to perform better than Supplementary Material
higher thresholds of 28% or 29% in 2 of the 3 good quality
studies making such comparisons.13,14,53 Supplementary material is available at Neuro-Oncology
To ensure fair comparison of methods, we assessed online.
comparisons on the same patients and tumors within a
study. A large variety of variants have been examined, par-
ticularly the use of different CpG sites and thresholds for
PSQ, as well as a mixture of use of FFPE and frozen tumor
Keywords
samples. There was only a small amount of direct replica- glioblastoma | meta-analysis | MGMT promoter methyla-
bility across studies, meaning that firm conclusions were tion | prognostic biomarker | temozolomide
difficult to draw.
We limited eligibility for the review to studies that re-
ported HRs or data sufficient for us to estimate them. In
many instances, we reconstructed time-to-event data from Acknowledgments
Kaplan-Meier curves, allowing us to include 14 studies that
we would not have included otherwise. However, there We thank Robin Grant (Co-ordinating Editor) and Gail Quinn
were still a small number of studies that had sought to (Managing Editor) for the Cochrane Gynaecological, Neuro-
compare methods but not presented data compatible with oncology and Orphan Cancers Group (GNOC) for their editorial
computation of HRs, which therefore did not meet our eli- guidance and patience. We also thank Joanne Platt (Information
gibility criteria. Specialist, GNOC) for aiding with the development of the search
We listed brief details of articles describing studies strategy. We also thank all of our external peer reviewers, in-
examining only 1 technique in the full Cochrane Review, cluding Karel Moons and Joie Ensor from the Cochrane
although these were not included in the final meta- Prognostic Methods Group, Catherine McBain, Andrew Bryant,
analysis (Table 4 and reference 12). Among the studies Helen Bulbeck, and Colin Smith.
that compared multiple techniques, we observed that HRs
1468 Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation

6. National Institute for Health and Care Excellence (NICE). Brain tumours
Funding (primary) and brain metastases in adults [NG99]. https://www.nice.org.
uk/guidance/ng99. Accessed June 2021.
This project was supported in part by the National Institute for 7. Hegi ME, Stupp R. Withholding temozolomide in glioblastoma patients
Health Research (NIHR), via Cochrane Programme Grant funding with unmethylated MGMT promoter—still a dilemma? Neuro Oncol.
(16/144) to the Cochrane Gynaecological, Neuro-oncology and 2015;17(11):1425–1427.
Orphan Cancers Group. A.M., F.S., and J.P.T.H. were supported in 8. Brandner S, von Deimling A. Diagnostic, prognostic and predictive rel-
part by Cancer Research United Kingdom (grant number C18281/ evance of molecular markers in gliomas. Neuropathol Appl Neurobiol.
A19169). S.B.  was partly supported by the NIHR Biomedical 2015;41(6):694–720.
Research Centre to University College London Hospitals. 9. Hegi  ME, Diserens  AC, Gorlia  T, et  al. MGMT gene silencing
and benefit from temozolomide in glioblastoma. N Engl J Med.
2005;352(10):997–1003.

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


10. Bady P, Sciuscio D, Diserens AC, et al. MGMT methylation analysis of
Conflict of interest statement. The views and opinions expressed glioblastoma on the Infinium methylation BeadChip identifies two dis-
herein are those of the review authors and do not necessarily tinct CpG regions associated with gene silencing and outcome, yielding
reflect those of the NIHR Systematic Reviews Programme, the a prediction model for comparisons across datasets, tumor grades, and
NIHR, the National Health Service (NHS), the Department of CIMP-status. Acta Neuropathol. 2012;124(4):547–560.
Health, or Cancer Research UK.
11. Capper  D, Jones  DTW, Sill  M, et  al. DNA methylation-based
classification of central nervous system tumours. Nature.
2018;555(7697):469–474.
12. McAleenan A, Kelly C, Spiga F, et al. Prognostic value of test(s) for O6-
Authorship statement. A.M., J.P.T.H., and K.M.K.  wrote the
protocol; S.D. designed and undertook the searches; K.M.K., methylguanine-DNA methyltransferase (MGMT) promoter methylation
C.L.F., S.B., and C.W.  provided a clinical perspective; A.M., for predicting overall survival in people with glioblastoma treated with
and K.M.K. screened search results; A.M., C.K., F.S., K.M.K., temozolomide. Cochrane Database Syst Rev. 2021;3:CD013316.
S.B., and C.L.F.  screened retrieved papers against eligibility 13. Quillien V, Lavenu A, Sanson M, et al. Outcome-based determination of
criteria; A.M., C.K., F.S., L.S., H.C., and J.P.T.H.  extracted optimal pyrosequencing assay for MGMT methylation detection in glio-
data from papers; A.M., C.K., F.S., L.S., and H.C.  appraised blastoma patients. J Neurooncol. 2014;116(3):487–496.
quality of included studies; J.P.T.H.  and H.C.  undertook the 14. Dunn J, Baborie A, Alam F, et al. Extent of MGMT promoter methylation
statistical analyses; J.P.T.H. undertook GRADE assessments; correlates with outcome in glioblastomas given temozolomide and radi-
K.M.K., S.B., J.P.T.H., F.S., and C.L.F.  interpreted the results; otherapy. Br J Cancer. 2009;101(1):124–131.
J.P.T.H.  and K.M.K.  co-ordinated the review; J.P.T.H.  wrote
15. Dahlrot  RH, Dowsett  J, Fosmark  S, et  al. Prognostic value of O-6-
up the full Cochrane Review with input from all authors; S.B.,
methylguanine-DNA methyltransferase (MGMT) protein expression in
K.M.K., and J.P.T.H. wrote this summary version.
glioblastoma excluding nontumour cells from the analysis. Neuropathol
Appl Neurobiol. 2018;44(2):172–184.
16. Håvik  AB, Brandal  P, Honne  H, et  al. MGMT promoter methylation in
gliomas-assessment by pyrosequencing and quantitative methylation-
specific PCR. J Transl Med. 2012;10:36.
17. Johannessen  LE, Brandal  P, Myklebust  TÅ, Heim  S, Micci  F,
References Panagopoulos  I. MGMT gene promoter methylation status - assess-
ment of two pyrosequencing kits and three methylation-specific PCR
methods for their predictive capacity in glioblastomas. Cancer Genomics
1. Louis  DN, Perry  A, Reifenberger  G, et  al. The 2016 World Health Proteomics. 2018;15(6):437–446.
Organization classification of tumors of the central nervous system: a 18. Hsu  CY, Ho  HL, Lin  SC, et  al. Prognosis of glioblastoma with
summary. Acta Neuropathol. 2016;131(6):803–820. faint MGMT methylation-specific PCR product. J Neurooncol.
2. Esteller M, Garcia-Foncillas J, Andion E, et al. Inactivation of the DNA- 2015;122(1):179–188.
repair gene MGMT and the clinical response of gliomas to alkylating 19. Hsu CY, Ho HL, Lin SC, et al. Comparative assessment of 4 methods to
agents. N Engl J Med. 2000;343(19):1350–1354. analyze MGMT status in a series of 121 glioblastoma patients. Appl
3. Malmström  A, Grønberg  BH, Marosi  C, et  al.; Nordic Clinical Brain Immunohistochem Mol Morphol. 2017;25(7):497–504.
Tumour Study Group (NCBTSG). Temozolomide versus standard 6-week 20. Karayan-Tapon  L, Quillien  V, Guilhot  J, et  al. Prognostic value
radiotherapy versus hypofractionated radiotherapy in patients older of O6-methylguanine-DNA methyltransferase status in glioblas-
than 60 years with glioblastoma: the Nordic randomised, phase 3 trial. toma patients, assessed by five different methods. J Neurooncol.
Lancet Oncol. 2012;13(9):916–926. 2010;97(3):311–322.
4. Wick  W, Platten  M, Meisner  C, et  al.; NOA-08 Study Group of 21. Kim DC, Kim KU, Kim YZ. Prognostic role of methylation status of the
Neuro-oncology Working Group (NOA) of German Cancer Society. MGMT promoter determined quantitatively by pyrosequencing in glio-
Temozolomide chemotherapy alone versus radiotherapy alone for malig- blastoma patients. J Korean Neurosurg Soc. 2016;59(1):26–36.
nant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. 22. Kristensen LS, Michaelsen SR, Dyrbye H, et al. Assessment of quanti-
Lancet Oncol. 2012;13(7):707–715. tative and allelic MGMT methylation patterns as a prognostic marker in
5. Weller M, van den Bent M, Tonn JC, et al.; European Association for glioblastoma. J Neuropathol Exp Neurol. 2016;75(3):246–255.
Neuro-Oncology (EANO) Task Force on Gliomas. European Association 23. McDonald KL, Rapkins RW, Olivier J, et al. The T genotype of the MGMT
for Neuro-Oncology (EANO) guideline on the diagnosis and treat- C>T (rs16906252) enhancer single-nucleotide polymorphism (SNP) is as-
ment of adult astrocytic and oligodendroglial gliomas. Lancet Oncol. sociated with promoter methylation and longer survival in glioblastoma
2017;18(6):e315–e329. patients. Eur J Cancer. 2013;49(2):360–368.
Brandner et al. Meta-analysis of the predictive value of MGMT promoter methylation 1469

Oncology
Neuro-
24. Quillien V, Lavenu A, Ducray F, et al. Validation of the high-performance 39. Lechapt-Zalcman E, Levallet G, Dugué AE, et al. O6-methylguanine-DNA
of pyrosequencing for clinical MGMT testing on a cohort of glioblastoma methyltransferase (MGMT) promoter methylation and low MGMT-
patients from a prospective dedicated multicentric trial. Oncotarget. encoded protein expression as prognostic markers in glioblastoma
2016;7(38):61916–61929. patients treated with biodegradable carmustine wafer implants after
25. Quillien  V, Lavenu  A, Ducray  F, et  al. Clinical validation of the CE-IVD initial surgery followed by radiotherapy with concomitant and adjuvant
marked Therascreen MGMT kit in a cohort of glioblastoma patients. temozolomide. Cancer. 2012;118(18):4545–4554.
Cancer Biomark. 2017;20(4):435–441. 40. Nguyen  A, Legrain  M, Noel  G, et  al. An innovative fluorescent semi-
26. Quillien  V, Lavenu  A, Karayan-Tapon  L, et  al. Comparative assess- quantitative methylation-specific PCR method for the determination of
ment of 5 methods (methylation-specific polymerase chain reaction, MGMT promoter methylation is reflecting intra-tumor heterogeneity.
MethyLight, pyrosequencing, methylation-sensitive high-resolution Curr Cancer Drug Targets. 2015;15(7):624–640.
melting, and immunohistochemistry) to analyze O6-methylguanine- 41. Bady  P, Sciuscio  D, Stupp  R, Delorenzi  M, Hegi  ME, Grp  EBT. MGMT
DNA-methyltranferase in a series of 100 glioblastoma patients. Cancer. methylation based outcome prediction is associated with two CpG re-

Downloaded from https://academic.oup.com/neuro-oncology/article/23/9/1457/6263562 by guest on 18 April 2023


2012;118(17):4201–4211. gions separated by a prediction minimum centred at the initiation start
27. Thon N, Thorsteinsdottir J, Eigenbrod S, et al. Outcome in unresectable site. Cancer Res. 2012;72(Suppl. 8):4031.
glioblastoma: MGMT promoter methylation makes the difference. J 42. Barault  L, Amatu  A, Bleeker  FE, et  al. Digital PCR quantification of
Neurol. 2017;264(2):350–358. MGMT methylation refines prediction of clinical benefit from alkylating
28. Barbagallo GM, Paratore S, Caltabiano R, et al. Long-term therapy with agents in glioblastoma and metastatic colorectal cancer. Ann Oncol.
temozolomide is a feasible option for newly diagnosed glioblastoma: a 2015;26(9):1994–1999.
single-institution experience with as many as 101 temozolomide cycles. 43. Binabaj MM, Bahrami A, ShahidSales S, et al. The prognostic value of
Neurosurg Focus. 2014;37(6):E4. MGMT promoter methylation in glioblastoma: a meta-analysis of clin-
29. Bell  EH, Pugh  SL, McElroy  JP, et  al. Molecular-based recursive ical trials. J Cell Physiol. 2018;233(1):378–386.
partitioning analysis model for glioblastoma in the temozolomide era: a 44. Zhao H, Wang S, Song C, Zha Y, Li L. The prognostic value of MGMT
correlative analysis based on NRG oncology RTOG 0525. JAMA Oncol. promoter status by pyrosequencing assay for glioblastoma patients’ sur-
2017;3(6):784–792. vival: a meta-analysis. World J Surg Oncol. 2016;14(1):261.
30. Felsberg  J, Thon  N, Eigenbrod  S, et  al.; German Glioma Network. 45. Zhang K, Wang XQ, Zhou B, Zhang L. The prognostic value of MGMT
Promoter methylation and expression of MGMT and the DNA mismatch promoter methylation in glioblastoma multiforme: a meta-analysis. Fam
repair genes MLH1, MSH2, MSH6 and PMS2 in paired primary and re- Cancer. 2013;12(3):449–458.
current glioblastomas. Int J Cancer. 2011;129(3):659–670. 46. EPPI-Reviewer: advanced software for systematic reviews, maps and
31. Lalezari  S, Chou  AP, Tran  A, et  al. Combined analysis of O6- evidence synthesis [computer program]. London: EPPI-Centre Software
methylguanine-DNA methyltransferase protein expression and promoter UCL Social Research Institute; 2020.
methylation provides optimized prognostication of glioblastoma out- 47. Guyot P, Ades AE, Ouwens MJ, Welton NJ. Enhanced secondary
come. Neuro Oncol. 2013;15(3):370–381. analysis of survival data: reconstructing the data from pub-
32. Lattanzio L, Borgognone M, Mocellini C, et al. MGMT promoter methyla- lished Kaplan-Meier survival curves. BMC Med Res Methodol.
tion and glioblastoma: a comparison of analytical methods and of tumor 2012;12:9.
specimens. Int J Biol Markers. 2015;30(2):e208–e216. 48. Hayden  JA, van  der  Windt  DA, Cartwright  JL, Côté  P, Bombardier  C.
33. Melguizo C, Prados J, González B, et al. MGMT promoter methylation Assessing bias in studies of prognostic factors. Ann Intern Med.
status and MGMT and CD133 immunohistochemical expression as prog- 2013;158(4):280–286.
nostic markers in glioblastoma patients treated with temozolomide plus 49. Deeks  JJ, Higgins  JPT, Altman  DG. Analysing data and under-
radiotherapy. J Transl Med. 2012;10:250. taking meta-analyses. In: Higgins  JPT, Thomas  J, Chandler  J, et  al.,
34. Park CK, Kim J, Yim SY, et al. Usefulness of MS-MLPA for detection of eds. Cochrane Handbook for Systematic Reviews of Interventions.
MGMT promoter methylation in the evaluation of pseudoprogression in Chichester: John Wiley & Sons; 2019:241–285.
glioblastoma patients. Neuro Oncol. 2011;13(2):195–202. 50. Guyatt GH, Oxman AD, Vist GE, et al.; GRADE Working Group. GRADE:
35. Yamashita S, Yokogami K, Matsumoto F, et al. MGMT promoter meth- an emerging consensus on rating quality of evidence and strength of re-
ylation in patients with glioblastoma: is methylation-sensitive high- commendations. BMJ. 2008;336(7650):924–926.
resolution melting superior to methylation-sensitive polymerase chain 51. Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG. Inactivation
reaction assay? J Neurosurg. 2018;130(3):780–788. of the DNA repair gene O6-methylguanine-DNA methyltransferase by
36. Yang  SH, Lee  KS, Yang  HJ, et  al. O6-methylguanine-DNA- promoter hypermethylation is a common event in primary human neo-
methyltransferase promoter methylation assessment by microdissection- plasia. Cancer Res. 1999;59(4):793–797.
assisted methylation-specific PCR and high resolution melting analysis in 52. Malley DS, Hamoudi RA, Kocialkowski S, Pearson DM, Collins VP,
patients with glioblastomas. J Neurooncol. 2012;106(2):243–250. Ichimura  K. A distinct region of the MGMT CpG island crit-
37. Yoshioka M, Matsutani T, Hara A, et al. Real-time methylation-specific ical for transcriptional regulation is preferentially methyl-
PCR for the evaluation of methylation status of MGMT gene in glioblas- ated in glioblastoma cells and xenografts. Acta Neuropathol.
toma. Oncotarget. 2018;9(45):27728–27735. 2011;121(5):651–661.
38. Felsberg J, Rapp M, Loeser S, et al. Prognostic significance of molecular 53. Brigliadori G, Foca F, Dall’Agata M, et al. Defining the cutoff value of
markers and extent of resection in primary glioblastoma patients. Clin MGMT gene promoter methylation and its predictive capacity in glio-
Cancer Res. 2009;15(21):6683–6693. blastoma. J Neurooncol. 2016;128(2):333–339.

You might also like