1
|
Satgunaseelan L, Lee M, Iannuzzi S, Hallal S, Deang K, Stanceski K, Wei H, Mason S, Shivalingam B, Sim HW, Buckland ME, Alexander KL. 'The Reports of My Death Are Greatly Exaggerated'-Evaluating the Effect of Necrosis on MGMT Promoter Methylation Testing in High-Grade Glioma. Cancers (Basel) 2024; 16:1906. [PMID: 38791984 PMCID: PMC11120496 DOI: 10.3390/cancers16101906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: MGMT (O-6-methylguanine-DNA methyltransferase) promoter methylation remains an important predictive biomarker in high-grade gliomas (HGGs). The influence of necrosis on the fidelity of MGMT promoter (MGMTp) hypermethylation testing is currently unknown. Therefore, our study aims to evaluate the effect of varying degrees of necrosis on MGMTp status, as determined by pyrosequencing, in a series of primary and recurrent HGGs; (2) Methods: Within each case, the most viable blocks (assigned as 'true' MGMTp status) and the most necrotic block were determined by histopathology review. MGMTp status was determined by pyrosequencing. Comparisons of MGMTp status were made between the most viable and most necrotic blocks. (3) Results: 163 samples from 64 patients with HGGs were analyzed. MGMTp status was maintained in 84.6% of primary and 78.3% of recurrent HGGs between the most viable and necrotic blocks. A threshold of ≥60% tumor cellularity was established at which MGMTp status was unaltered, irrespective of the degree of necrosis. (4) Conclusions: MGMTp methylation status, as determined by pyrosequencing, does not appear to be influenced by necrosis in the majority of cases at a cellularity of at least 60%. Further investigation into the role of intratumoral heterogeneity on MGMTp status will increase our understanding of this predictive marker.
Collapse
Affiliation(s)
- Laveniya Satgunaseelan
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Maggie Lee
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Sebastian Iannuzzi
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Susannah Hallal
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
| | - Kristine Deang
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
| | - Kristian Stanceski
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Heng Wei
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Sofia Mason
- Department of Medical Oncology, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (S.M.); (H.-W.S.)
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Brindha Shivalingam
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
- Department of Neurosurgery, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Hao-Wen Sim
- Department of Medical Oncology, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (S.M.); (H.-W.S.)
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW 2050, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Darlinghurst, NSW 2010, Australia
| | - Michael E. Buckland
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Kimberley L. Alexander
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
| |
Collapse
|
2
|
Li M, Dong G, Zhang W, Ren X, Jiang H, Yang C, Zhao X, Zhu Q, Li M, Chen H, Yu K, Cui Y, Song L. Combining MGMT promoter pyrosequencing and protein expression to optimize prognosis stratification in glioblastoma. Cancer Sci 2021; 112:3699-3710. [PMID: 34115910 PMCID: PMC8409410 DOI: 10.1111/cas.15024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/16/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Pyrosequencing (PSQ) represents the golden standard for MGMT promoter status determination. Binary interpretation of results based on the threshold from the average of several CpGs tested would neglect the existence of the “gray zone”. How to define the gray zone and reclassify patients in this subgroup remains to be elucidated. A consecutive cohort of 312 primary glioblastoma patients were enrolled. CpGs 74‐81 in the promoter region of MGMT were tested by PSQ and the protein expression was assessed by immunohistochemistry (IHC). Receiver operating characteristic curves were constructed to calculate the area under the curves (AUC). Kaplan‐Meier plots were used to estimate the survival rate of patients compared by the log‐rank test. The optimal threshold of each individual CpG differed from 5% to 11%. Patients could be separated into the hypomethylated subgroup (all CpGs tested below the corresponding optimal thresholds, n = 126, 40.4%), hypermethylated subgroup (all CpGs tested above the corresponding optimal thresholds, n = 108, 34.6%), and the gray zone subgroup (remaining patients, n = 78, 25.0%). Patients in the gray zone harbored an intermediate prognosis. The IHC score instead of the average methylation levels could successfully predict the prognosis for the gray zone (AUC for overall survival, 0.653 and 0.519, respectively). Combining PSQ and IHC significantly improved the efficiency of survival prediction (AUC: 0.662, 0.648, and 0.720 for PSQ, IHC, and combined, respectively). Immunohistochemistry is a robust method to predict prognosis for patients in the gray zone defined by PSQ. Combining PSQ and IHC could significantly improve the predictive ability for clinical outcomes.
Collapse
Affiliation(s)
- Mingxiao Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Gehong Dong
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weiwei Zhang
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaohui Ren
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Haihui Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chuanwei Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xuzhe Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qinghui Zhu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ming Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hongyan Chen
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kefu Yu
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yong Cui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lin Song
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Institute for Brain Disorders and Beijing Key Laboratory of Brain Tumor, Beijing, China.,Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Mansouri A, Hachem LD, Mansouri S, Nassiri F, Laperriere NJ, Xia D, Lindeman NI, Wen PY, Chakravarti A, Mehta MP, Hegi ME, Stupp R, Aldape KD, Zadeh G. MGMT promoter methylation status testing to guide therapy for glioblastoma: refining the approach based on emerging evidence and current challenges. Neuro Oncol 2020; 21:167-178. [PMID: 30189035 DOI: 10.1093/neuonc/noy132] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/11/2018] [Accepted: 08/29/2018] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor, with a universally poor prognosis. The emergence of molecular biomarkers has had a significant impact on histological typing and diagnosis, as well as predicting patient survival and response to treatment. The methylation status of the O6-methylguanine-DNA methyl-transferase (MGMT) gene promoter is one such molecular biomarker. Despite the strong evidence supporting the role of MGMT methylation status in prognostication, its routine implementation in clinical practice has been challenging. The methods and optimal cutoff definitions for MGMT status determination remain controversial. Variation in detection methods between laboratories presents a major challenge for consensus. Moreover, consideration of other clinical and genetic/epigenetic factors must also be incorporated into treatment decision making. In this review, we distill the available evidence to summarize our position on the optimal use of available assays, and propose strategies for resolving cases with equivocal methylation status and a framework for incorporating this important assay into research and clinical practice.
Collapse
Affiliation(s)
- Alireza Mansouri
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Laureen D Hachem
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Sheila Mansouri
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Farshad Nassiri
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Normand J Laperriere
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Daniel Xia
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Arnab Chakravarti
- Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Miami, Florida, USA
| | - Monika E Hegi
- Department of Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland
| | - Roger Stupp
- Malnati Brain Tumor Institute of the Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kenneth D Aldape
- Department of Laboratory Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Gelareh Zadeh
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Abstract
Gliomas, that do not respond to alkylating agent chemotherapy, can be made more sensitive to chemotherapy through promotor mediated epigenetic silencing of the MGMT gene. MGMT is one of the important markers in glioblastomas as it not only predicts response to therapy but may also be used as an independent prognostic marker. As such, MGMT is gaining increasing traction in diagnosis, prognostication, and therapeutic decision-making for these highly malignant gliomas. Although, MGMT promotor methylation status is becoming more commonly used in neuro-oncology; this test remains imperfect. Because of its increasing use in clinical practice and research, it is integral that we are aware of its pitfalls and complications. Currently, there are many ways to detect a patient's MGMT promotor methylation status, including: quantitative PCR, methylation-specific PCR, pyrosequencing, real time PCR with high resolution melt, and the infinitum methylation EPIC beadChip. The technical aspects, shortcomings, and optimal approach to interpreting the results of each method will be discussed. Furthermore, given that none of these methods have been prospectively validated, the challenge of equivocal cases will be discussed, and technical and logistic strategies for overcoming these challenges will be proposed. Finally, the difficulty in validating these methods, establishing standardized practice, and considerations of the cost of these competing methods will be explored.
Collapse
|
5
|
Braczynski AK, Capper D, Jones DTW, Schittenhelm J, Stichel D, von Deimling A, Harter PN, Mittelbronn M. High density DNA methylation array is a reliable alternative for PCR-based analysis of the MGMT promoter methylation status in glioblastoma. Pathol Res Pract 2019; 216:152728. [PMID: 31784096 DOI: 10.1016/j.prp.2019.152728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/08/2019] [Accepted: 11/10/2019] [Indexed: 10/25/2022]
Abstract
AIM MGMT promoter methylation status is an important biomarker predicting survival and response to chemotherapy in patients suffering from glioblastoma. Since new diagnostic methods such as methylome-based classification of brain tumors are more and more frequently performed, we aimed at comparing the suitability of calculating the MGMT promoter methylation status in a quantitative manner from the methylome profiling as compared to the classic gold standard assessment by PCR. METHODS Our cohort consisted of 39 cases diagnosed as "glioblastoma, IDH-wildtype" of which the MGMT promoter methylation status was analyzed with both methylation-specific PCR and high density DNA methylation array using the STP-27 algorithm. Contradictory results were validated by pyrosequencing. RESULTS The inter-method reliability reached 77% (kappa-coefficient: 0.58) when also cases with an inconclusive result in one or the other method were taken into account. When only cases with conclusive results in both methods were considered, a very high inter-method reliability of 91% (kappa-coefficient: 0.86) could be achieved. For "methylated" cases, no contradictory results were obtained. For the remaining two cases with discrepant results subsequent pyrosequencing analyses spoke in favor of each previously applied method once. CONCLUSION In addition to its benefits for molecular subgrouping and copy number analysis of brain tumors, DNA-methylation based classification is a highly reliable tool for the assessment of MGMT promoter methylation status in glioblastoma patients.
Collapse
Affiliation(s)
- Anne K Braczynski
- Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany; Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany
| | - David Capper
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neuropathology, Charité Universitätsmedizin Berlin and German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - David T W Jones
- Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Jens Schittenhelm
- Department of Neuropathology, Institute of Pathology and Neuropathology, Eberhard-Karls University and Comprehensive Cancer Center Tuebingen-Stuttgart, Tuebingen, Germany
| | - Damian Stichel
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick N Harter
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
| | - Michel Mittelbronn
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany; NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health (LIH), Luxembourg; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg; National Center of Pathology (NCP), Laboratoire national de santé (LNS), Dudelange, Luxembourg; Luxembourg Centre of Neuropathology (LCNP), Luxembourg.
| |
Collapse
|
6
|
Weak MGMT gene promoter methylation confers a clinically significant survival benefit in patients with newly diagnosed glioblastoma: a retrospective cohort study. J Neurooncol 2019; 146:55-62. [DOI: 10.1007/s11060-019-03334-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 11/03/2019] [Indexed: 12/25/2022]
|
7
|
A novel analytical model of MGMT methylation pyrosequencing offers improved predictive performance in patients with gliomas. Mod Pathol 2019; 32:4-15. [PMID: 30291347 DOI: 10.1038/s41379-018-0143-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/29/2018] [Accepted: 08/31/2018] [Indexed: 12/28/2022]
Abstract
The methylation status of the promoter of MGMT gene is a crucial factor influencing clinical decision-making in patients with gliomas. MGMT pyrosequencing results are often dichotomized by a cut-off value based on an average of several tested CpGs. However, this method frequently results in a "gray zone", representing a dilemma for physicians. We therefore propose a novel analytical model for MGMT methylation pyrosequencing. MGMT CpG heterogeneity was investigated in 213 glioma patients in two tested cohorts: cohort A in which CpGs 75-82 were tested and cohort B in which CpGs 72-78 were tested. The predictive performances of the novel and traditional averaging models were compared in 135 patients who received temozolomide using receiver operating characteristic curves and Kaplan-Meier curves, and in patients stratified according to isocitrate dehydrogenase gene mutation status. The results were validated in an independent cohort of 65 consecutive patients with high-grade gliomas from the Chinese Glioma Genome Atlas database. Heterogeneity of MGMT promoter CpG methylation level was observed in most gliomas. The optimal cut-off value for each individual CpG varied from 4-16%. The current analysis defined MGMT promoter methylation as occurring when at least three CpGs exceeded their respective cut-off values. This novel analysis could accurately predict the prognosis of patients in the methylation "gray zone" according to the standard averaging method, and improved the area under the curves from 0.67, 0.76, and 0.67 to 0.70, 0.84, and 0.72 in cohorts A, B, and the validation cohort, respectively, demonstrating superiority of this analytical method in all three cohorts. Furthermore, the advantages of the novel analysis were retained regardless of WHO grade and isocitrate dehydrogenase gene mutation status. In conclusion, this novel analytical model offers an improved clinical predictive performance for MGMT pyrosequencing results and is suitable for clinical use in patients with gliomas.
Collapse
|
8
|
Hegi ME, Genbrugge E, Gorlia T, Stupp R, Gilbert MR, Chinot OL, Nabors LB, Jones G, Van Criekinge W, Straub J, Weller M. MGMT Promoter Methylation Cutoff with Safety Margin for Selecting Glioblastoma Patients into Trials Omitting Temozolomide: A Pooled Analysis of Four Clinical Trials. Clin Cancer Res 2018; 25:1809-1816. [PMID: 30514777 DOI: 10.1158/1078-0432.ccr-18-3181] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/26/2018] [Accepted: 11/29/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE The methylation status of the O6-methylguanine DNA methyltransferase (MGMT) gene promoter is predictive for benefit from temozolomide in glioblastoma (GBM). A clinically optimized cutoff was sought allowing patient selection for therapy without temozolomide, while avoiding to withhold it from patients who may potentially benefit.Experimental Design: Quantitative MGMT methylation-specific PCR data were obtained for newly diagnosed patients with GBM screened or treated with standard radiotherapy and temozolomide in four randomized trials. The pooled dataset was randomly split into a training and test dataset. The unsupervised cutoff was obtained at a 50% probability to be (un)methylated. ROC analysis identified an optimal cutoff supervised by overall survival (OS). RESULTS For 4,041 patients valid MGMT results were obtained, whereof 1,725 were randomized. The unsupervised cutoff in the training dataset was 1.27 (log2[1,000 × (MGMT+1)/ACTB]), separating unmethylated and methylated patients. The optimal supervised cutoff for unmethylated patients was -0.28 (AUC = 0.61), classifying "truly unmethylated" (≤-0.28) and "gray zone" patients (>-0.28, ≤1.27), the latter comprising approximately 10% of cases. In contrast, for patients with MGMT methylation (>1.27) more methylation was not related to better outcome. Both methylated and gray zone patients performed significantly better for OS than truly unmethylated patients [HR = 0.35, 95% confidence interval (CI), 0.27-0.45, P < 0.0001; HR = 0.58, 95% CI, 0.43-0.78, P < 0.001], validated in the test dataset. The MGMT assay was highly reproducible upon retesting of 218 paired samples (R 2 = 0.94). CONCLUSIONS Low MGMT methylation (gray zone) may confer some sensitivity to temozolomide treatment, hence the lower safety margin should be considered for selecting patients with unmethylated GBM into trials omitting temozolomide.
Collapse
Affiliation(s)
- Monika E Hegi
- Neurosurgery & Neuroscience Research Center, Lausanne University Hospital, Lausanne, Switzerland.
| | - Els Genbrugge
- European Organisation for Treatment and Research of Cancer (EORTC) Data Centre, Brussels, Belgium
| | - Thierry Gorlia
- European Organisation for Treatment and Research of Cancer (EORTC) Data Centre, Brussels, Belgium
| | - Roger Stupp
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Olivier L Chinot
- Aix-Marseille University, AP-HM, Hôpital de la Timone, Marseille, France
| | - L Burt Nabors
- University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Wim Van Criekinge
- Department of Mathematical Modeling, Statistics and Bio-Informatics, Ghent University, Ghent, Belgium
| | | | - Michael Weller
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Tam BE, Hao Y, Sikes HD. An examination of critical parameters in hybridization-based epigenotyping using magnetic microparticles. Biotechnol Prog 2018; 34:1589-1595. [PMID: 29693329 DOI: 10.1002/btpr.2644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 04/14/2018] [Indexed: 12/24/2022]
Abstract
Gene-specific promoter methylation is involved in gene silencing and is an important cancer biomarker. Cancer-specific methylation patterns have been observed and clinically validated for numerous gene promoters, but the knowledge gleaned from this large body of work is currently under-utilized in the clinic. Methylation-specific PCR is currently the gold standard method for clinical methylation assessment, but several research groups have proposed hybridization-based techniques which could be simpler to implement and provide more accurate results. However, the sensitivity of this easier alternative must be improved dramatically in order to compete with methylation-specific PCR. Efficient sample capture is a key step in maximizing sensitivity, so here we investigate the key parameters involved in (i) maximizing the capture of gene-specific target DNA molecules at the surfaces of functionalized, magnetic microparticles and (ii) recognizing DNA methylation using an engineered methyl-CpG-binding domain (MBD) protein. The magnetic bead density, the probe concentration, and the MBD concentration were very important for maximizing detection, and other variables such as the hybridization time also impacted the target capture efficiency but had a smaller effect on the overall methylation assay. The effect of genomic DNA on the capture of the target sequence was also investigated, and model methylated vs. unmethylated target sequences could be distinguished in the presence of 1 ng/μL genomic DNA. The findings we report related to the underlying binding events involved in hybridization-based epigenotyping can be leveraged in combination with the many signal amplification and detection approaches that are currently being developed. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:1589-1595, 2018.
Collapse
Affiliation(s)
- Brooke E Tam
- Dept. of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139
| | - Yining Hao
- Dept. of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139
| | - Hadley D Sikes
- Dept. of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139
| |
Collapse
|
10
|
Kameda-Smith MM, Manoranjan B, Bakhshinyan D, Adile AA, Venugopal C, Singh SK. Brain tumor initiating cells: with great technology will come greater understanding. FUTURE NEUROLOGY 2017. [DOI: 10.2217/fnl-2017-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The discovery of the brain tumor initiating cells resulted in a paradigm shift within the cancer research community to consider brain tumors as an outcome of developmental mechanisms gone awry. This review will guide the reader through the technological advances that hold the powerful potential to allow brain cancer researchers to develop an intimate understanding of the dynamic and complex mechanism governing brain tumor behavior.
Collapse
Affiliation(s)
- Michelle M Kameda-Smith
- Stem Cell & Cancer Research Institute (SCC-RI), McMaster University, Michael DeGroote Center for Learning & Discovery, Room 5061, 1200 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
- Division of Neurosurgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Branavan Manoranjan
- Stem Cell & Cancer Research Institute (SCC-RI), McMaster University, Michael DeGroote Center for Learning & Discovery, Room 5061, 1200 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
| | - David Bakhshinyan
- Stem Cell & Cancer Research Institute (SCC-RI), McMaster University, Michael DeGroote Center for Learning & Discovery, Room 5061, 1200 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
| | - Ashley A Adile
- Stem Cell & Cancer Research Institute (SCC-RI), McMaster University, Michael DeGroote Center for Learning & Discovery, Room 5061, 1200 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
| | - Chitra Venugopal
- Stem Cell & Cancer Research Institute (SCC-RI), McMaster University, Michael DeGroote Center for Learning & Discovery, Room 5061, 1200 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
| | - Sheila K Singh
- Stem Cell & Cancer Research Institute (SCC-RI), McMaster University, Michael DeGroote Center for Learning & Discovery, Room 5061, 1200 Main Street West, Hamilton, Ontario, L8S 4K1, Canada
- Division of Neurosurgery, Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
11
|
Ida CM, Butz ML, Jenkins RB, Sarkaria JN, Kitange GJ, Giannini C, Kipp BR. Real-Time Methylation-Specific Polymerase Chain Reaction for MGMT Promoter Methylation Clinical Testing in Glioblastoma: An Alternative Detection Method for a Heterogeneous Process. Am J Clin Pathol 2017; 148:296-307. [PMID: 28967952 DOI: 10.1093/ajcp/aqx073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES To develop and evaluate a real-time methylation-specific polymerase chain reaction (RT-MSP) MGMT assay, with a particular focus on small biopsies and indeterminate testing results. METHODS We assessed formalin-fixed paraffin-embedded glioblastoma or gliosarcoma specimens (n = 641). A test-validation group (n = 51) with previously obtained reference laboratory (RL) results was used to determine performance characteristics of the RT-MSP assay. An indeterminate (equivocal) category was established for cases that could not be clearly classified as positive or negative. RESULTS Overall agreement of RT-MSP and RL results was 91% (41/45 nonindeterminate cases). Discordant cases were tested by pyrosequencing, and results were most concordant with RT-MSP. Among cases with limited amounts of tissue (n = 7), six yielded valid results by RT-MSP (all negative); the single invalid result consisted of a stereotactic biopsy specimen obtained 14 years prior. A subset of indeterminate cases obtained during clinical testing (n = 18/575 [3%]) was also evaluated by pyrosequencing and showed a heterogeneous pattern of methylation across the eight interrogated CpG sites. CONCLUSIONS The RT-MSP assay that we developed in-house is a robust clinical detection method for the heterogeneous process of MGMT promoter methylation in glioblastoma.
Collapse
Affiliation(s)
| | | | - Robert B Jenkins
- Departments of Laboratory Medicine and Pathology
- Biochemistry and Molecular Biology
| | | | | | | | - Benjamin R Kipp
- Departments of Laboratory Medicine and Pathology
- Clinical Genomics
| |
Collapse
|
12
|
Novel recursive partitioning analysis classification for newly diagnosed glioblastoma: A multi-institutional study highlighting the MGMT promoter methylation and IDH1 gene mutation status. Radiother Oncol 2017; 123:106-111. [PMID: 28302331 DOI: 10.1016/j.radonc.2017.02.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND PURPOSE To refine the recursive partitioning analysis (RPA) classification for glioblastoma incorporating the MGMT methylation and IDH1 mutation status. METHODS AND MATERIALS Three-hundred forty patients were treated with radiotherapy plus concurrent and adjuvant temozolomide in three tertiary-referral hospitals. MGMT methylation and IDH1 mutation status were available in all patients. Methylation of the MGMT (MGMTmeth) and mutation of IDH1 (IDH1mut) were observed in 42.4% and 6.2% of the patients, respectively. RESULTS The median follow-up for survivors and all patients was 33.2 and 20.5months, respectively. The median survival (MS) was 23.6months. RPA was performed on behalf of the results of the Cox proportional hazards model. MGMT methylation generated the initial partition (MGMTmeth vs. MGMTunmeth) in the RPA. Three final RPA classes were identified; class I=MGMTmeth/IDH1mut or MGMTmeth/IDH1wt/GTR/KPS≥90 (MS, 67.2months); class II=MGMTmeth/IDH1wt/GTR/KPS<90, MGMTmeth/IDH1wt/residual disease, MGMTunmeth/age<50, or MGMTunmeth/age≥50/GTR (MS, 24.0months); class III=MGMTunmeth/age≥50/residual disease (MS, 15.2months). CONCLUSIONS A novel RPA classification for glioblastoma was formulated highlighting the impact of MGMTmeth and IDH1mut in the temozolomide era. This model integrating pertinent molecular information can be used effectively for the patient stratification in future clinical trials. An external validation is ongoing.
Collapse
|