1
|
Zappe K, Pühringer K, Pflug S, Berger D, Weis S, Spiegl-Kreinecker S, Cichna-Markl M. Association of MGMT Promoter and Enhancer Methylation with Genetic Variants, Clinical Parameters, and Demographic Characteristics in Glioblastoma. Cancers (Basel) 2023; 15:5777. [PMID: 38136323 PMCID: PMC10742072 DOI: 10.3390/cancers15245777] [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: 11/03/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
The response of glioblastoma (GBM) patients to the alkylating agent temozolomide (TMZ) vitally depends on the expression level of the repair protein O6-methylguanine-DNA methyltransferase (MGMT). Since MGMT is strongly regulated by promoter methylation, the methylation status of the MGMT promoter has emerged as a prognostic and predictive biomarker for GBM patients. By determining the methylation levels of the four enhancers located within or close to the MGMT gene, we recently found that enhancer methylation contributes to MGMT regulation. In this study, we investigated if methylation of the four enhancers is associated with SNP rs16906252, TERT promoter mutations C228T and C250T, TERT SNP rs2853669, proliferation index Ki-67, overall survival (OS), age, and sex of the patients. In general, associations with genetic variants, clinical parameters, and demographic characteristics were caused by a complex interplay of multiple CpGs in the MGMT promoter and of multiple CpGs in enhancer regions. The observed associations for intragenic enhancer 4, located in intron 2 of MGMT, differed from associations observed for the three intergenic enhancers. Some findings were restricted to subgroups of samples with either methylated or unmethylated MGMT promoters, underpinning the relevance of the MGMT promoter status in GBMs.
Collapse
Affiliation(s)
- Katja Zappe
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (K.Z.); (K.P.); (S.P.); (D.B.)
| | - Katharina Pühringer
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (K.Z.); (K.P.); (S.P.); (D.B.)
| | - Simon Pflug
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (K.Z.); (K.P.); (S.P.); (D.B.)
| | - Daniel Berger
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (K.Z.); (K.P.); (S.P.); (D.B.)
| | - Serge Weis
- Division of Neuropathology, Department of Pathology and Molecular Pathology, Kepler University Hospital GmbH, Johannes Kepler University, 4040 Linz, Austria;
| | - Sabine Spiegl-Kreinecker
- Department of Neurosurgery, Kepler University Hospital GmbH, Johannes Kepler University, 4040 Linz, Austria;
| | - Margit Cichna-Markl
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; (K.Z.); (K.P.); (S.P.); (D.B.)
| |
Collapse
|
2
|
Sim HW, Wachsmuth L, Barnes EH, Yip S, Koh ES, Hall M, Jennens R, Ashley DM, Verhaak RG, Heimberger AB, Rosenthal MA, Hovey EJ, Ellingson BM, Tognela A, Gan HK, Wheeler H, Back M, McDonald KL, Long A, Cuff K, Begbie S, Gedye C, Mislang A, Le H, Johnson MO, Kong BY, Simes JR, Lwin Z, Khasraw M. NUTMEG: A randomized phase II study of nivolumab and temozolomide versus temozolomide alone in newly diagnosed older patients with glioblastoma. Neurooncol Adv 2023; 5:vdad124. [PMID: 37841696 PMCID: PMC10576515 DOI: 10.1093/noajnl/vdad124] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
Background There is an immunologic rationale to evaluate immunotherapy in the older glioblastoma population, who have been underrepresented in prior trials. The NUTMEG study evaluated the combination of nivolumab and temozolomide in patients with glioblastoma aged 65 years and older. Methods NUTMEG was a multicenter 2:1 randomized phase II trial for patients with newly diagnosed glioblastoma aged 65 years and older. The experimental arm consisted of hypofractionated chemoradiation with temozolomide, then adjuvant nivolumab and temozolomide. The standard arm consisted of hypofractionated chemoradiation with temozolomide, then adjuvant temozolomide. The primary objective was to improve overall survival (OS) in the experimental arm. Results A total of 103 participants were randomized, with 69 in the experimental arm and 34 in the standard arm. The median (range) age was 73 (65-88) years. After 37 months of follow-up, the median OS was 11.6 months (95% CI, 9.7-13.4) in the experimental arm and 11.8 months (95% CI, 8.3-14.8) in the standard arm. For the experimental arm relative to the standard arm, the OS hazard ratio was 0.85 (95% CI, 0.54-1.33). In the experimental arm, there were three grade 3 immune-related adverse events which resolved, with no unexpected serious adverse events. Conclusions Due to insufficient evidence of benefit with nivolumab, the decision was made not to transition to a phase III trial. No new safety signals were identified with nivolumab. This complements the existing series of immunotherapy trials. Research is needed to identify biomarkers and new strategies including combinations.
Collapse
Affiliation(s)
- Hao-Wen Sim
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Department of Medical Oncology, Chris O’Brien Lifehouse, Sydney, New South Wales, Australia
| | - Luke Wachsmuth
- The Brain Tumor Immunotherapy Program, Duke University School of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Elizabeth H Barnes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Sonia Yip
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Eng-Siew Koh
- Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Radiation Oncology, Liverpool Hospital, Sydney, New South Wales, Australia
| | - Merryn Hall
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Ross Jennens
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Epworth HealthCare Richmond, Melbourne, Victoria, Australia
| | - David M Ashley
- The Preston Robert Tisch Brain Tumor Center, Duke University School of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Roel G Verhaak
- The Jackson Laboratory for Genomic Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Amy B Heimberger
- Department of Neurological Surgery, Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mark A Rosenthal
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Elizabeth J Hovey
- Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Medical Oncology, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, University of California Los Angeles, Los Angeles, California, USA
| | - Annette Tognela
- Department of Medical Oncology, Campbelltown Hospital, Sydney, New South Wales, Australia
| | - Hui K Gan
- Department of Medical Oncology, Austin Hospital, Melbourne, Victoria, Australia
| | - Helen Wheeler
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Michael Back
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Kerrie L McDonald
- Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Anne Long
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Katharine Cuff
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Stephen Begbie
- Department of Medical Oncology, Port Macquarie Base Hospital, Port Macquarie, New South Wales, Australia
| | - Craig Gedye
- Department of Medical Oncology, Calvary Mater Newcastle, Newcastle, New South Wales, Australia
| | - Anna Mislang
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
- Department of Medical Oncology, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Hien Le
- Department of Radiation Oncology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Margaret O Johnson
- The Preston Robert Tisch Brain Tumor Center, Duke University School of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Benjamin Y Kong
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Medical Oncology, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - John R Simes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
- Department of Medical Oncology, Chris O’Brien Lifehouse, Sydney, New South Wales, Australia
| | - Zarnie Lwin
- School of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Department of Medical Oncology, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
| | - Mustafa Khasraw
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
- The Brain Tumor Immunotherapy Program, Duke University School of Medicine, Duke University Medical Center, Durham, North Carolina, USA
- The Preston Robert Tisch Brain Tumor Center, Duke University School of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
3
|
Tian Y, Liu H, Zhang C, Liu W, Wu T, Yang X, Zhao J, Sun Y. Comprehensive Analyses of Ferroptosis-Related Alterations and Their Prognostic Significance in Glioblastoma. Front Mol Biosci 2022; 9:904098. [PMID: 35720126 PMCID: PMC9204216 DOI: 10.3389/fmolb.2022.904098] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/27/2022] [Indexed: 12/23/2022] Open
Abstract
Background: This study was designed to explore the implications of ferroptosis-related alterations in glioblastoma patients.Method: After obtaining the data sets CGGA325, CGGA623, TCGA-GBM, and GSE83300 online, extensive analysis and mutual verification were performed using R language-based analytic technology, followed by further immunohistochemistry staining verification utilizing clinical pathological tissues.Results: The analysis revealed a substantial difference in the expression of ferroptosis-related genes between malignant and paracancerous samples, which was compatible with immunohistochemistry staining results from clinicopathological samples. Three distinct clustering studies were run sequentially on these data. All of the findings were consistent and had a high prediction value for glioblastoma. Then, the risk score predicting model containing 23 genes (CP, EMP1, AKR1C1, FMOD, MYBPH, IFI30, SRPX2, PDLIM1, MMP19, SPOCD1, FCGBP, NAMPT, SLC11A1, S100A10, TNC, CSMD3, ATP1A2, CUX2, GALNT9, TNFAIP6, C15orf48, WSCD2, and CBLN1) on the basis of “Ferroptosis.gene.cluster” was constructed. In the subsequent correlation analysis of clinical characteristics, tumor mutation burden, HRD, neoantigen burden and chromosomal instability, mRNAsi, TIDE, and GDSC, all the results indicated that the risk score model might have a better predictive efficiency.Conclusion: In glioblastoma, there were a large number of abnormal ferroptosis-related alterations, which were significant for the prognosis of patients. The risk score-predicting model integrating 23 genes would have a higher predictive value.
Collapse
Affiliation(s)
- Yuan Tian
- Somatic Radiotherapy Department, Shandong Second Provincial General Hospital, Jinan, China
- *Correspondence: Yuan Tian, ; Yuping Sun,
| | - Hongtao Liu
- Department of Pathology, Shandong Medicine and Health Key Laboratory of Clinical Pathology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Institute of Nephrology, Jinan, China
| | - Caiqing Zhang
- Department of Respiratory and Critical Care Medicine, Shandong Second Provincial General Hospital, Shandong University, Jinan, China
| | - Wei Liu
- Somatic Radiotherapy Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Tong Wu
- Somatic Radiotherapy Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Xiaowei Yang
- Department of Hepatobiliary Intervention, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Junyan Zhao
- Nursing Department, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yuping Sun
- Phase I Clinical Trial Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- *Correspondence: Yuan Tian, ; Yuping Sun,
| |
Collapse
|
4
|
Caccese M, Simonelli M, Villani V, Rizzato S, Ius T, Pasqualetti F, Russo M, Rudà R, Amoroso R, Bellu L, Bertorelle R, Cavallin F, Dipasquale A, Carosi M, Pizzolitto S, Cesselli D, Persico P, Casini B, Fassan M, Zagonel V, Lombardi G. Definition of the Prognostic Role of MGMT Promoter Methylation Value by Pyrosequencing in Newly Diagnosed IDH Wild-Type Glioblastoma Patients Treated with Radiochemotherapy: A Large Multicenter Study. Cancers (Basel) 2022; 14:2425. [PMID: 35626029 PMCID: PMC9139569 DOI: 10.3390/cancers14102425] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Background. O6-methylguanine (O6-MeG)-DNA methyltransferase (MGMT) methylation status is a predictive factor for alkylating treatment efficacy in glioblastoma patients, but its prognostic role is still unclear. We performed a large, multicenter study to evaluate the association between MGMT methylation value and survival. Methods. We evaluated glioblastoma patients with an assessment of MGMT methylation status by pyrosequencing from nine Italian centers. The inclusion criteria were histological diagnosis of IDH wild-type glioblastoma, Eastern Cooperative Oncology Group Performance Status (ECOG-PS) ≤2, and radio-chemotherapy treatment with temozolomide. The relationship between OS and MGMT was investigated with a time-dependent Receiver Operating Characteristics (ROC) curve and Cox regression models. Results. In total, 591 newly diagnosed glioblastoma patients were analyzed. The median OS was 16.2 months. The ROC analysis suggested a cut-off of 15% for MGMT methylation. The 2-year Overall Survival (OS) was 18.3% and 51.8% for MGMT methylation <15% and ≥15% (p < 0.0001). In the multivariable analysis, MGMT methylation <15% was associated with impaired survival (p < 0.00001). However, we also found a non-linear association between MGMT methylation and OS (p = 0.002): median OS was 14.8 months for MGMT in 0−4%, 18.9 months for MGMT in 4−40%, and 29.9 months for MGMT in 40−100%. Conclusions. Our findings suggested a non-linear relationship between OS and MGMT promoter methylation, which implies a varying magnitude of prognostic effect across values of MGMT promoter methylation by pyrosequencing in newly diagnosed IDH wild-type glioblastoma patients treated with chemoradiotherapy.
Collapse
Affiliation(s)
- Mario Caccese
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (V.Z.); (G.L.)
| | - Matteo Simonelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (M.S.); (A.D.); (P.P.)
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Veronica Villani
- Neuro-Oncology Unit, Regina Elena National Cancer Institute, 00161 Rome, Italy;
| | - Simona Rizzato
- Department of Oncology, Central Friuli University Health Authority, 33100 Udine, Italy;
| | - Tamara Ius
- Neurosurgery Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy;
| | - Francesco Pasqualetti
- Radiation Oncology Unit, Pisa University Hospital, 56121 Pisa, Italy;
- Department of Oncology, University of Oxford, Oxford OX1 4BH, UK
| | - Marco Russo
- Neurology Unit, Neuromotor Department, Azienda USL-IRCCS Reggio Emilia, 42121 Emilia, Italy;
| | - Roberta Rudà
- Department of Neuro-Oncology, University of Turin and City of Health and Science Hospital, 10094 Torino, Italy;
- Neurology Unit, Hospital of Castelfranco Veneto, 31033 Castelfranco Veneto, Italy
| | - Rosina Amoroso
- Neurosurgery Unit, Department of Surgery, Hospital of Livorno, Azienda Asl Toscana Nord Ovest, 57100 Livorno, Italy;
| | - Luisa Bellu
- Radiotherapy and Radiosurgery Department, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy;
| | - Roberta Bertorelle
- Immunology and Molecular Oncology Unit, Department of Oncology, Veneto Institute of Oncology IOV IRCCS, 35128 Padua, Italy;
| | | | - Angelo Dipasquale
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (M.S.); (A.D.); (P.P.)
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Mariantonia Carosi
- Pathology Unit, Regina Elena National Cancer Institute, 00161 Rome, Italy; (M.C.); (B.C.)
| | - Stefano Pizzolitto
- Department of Surgical Pathology, Central Friuli University Health Authority, 33100 Udine, Italy;
| | - Daniela Cesselli
- Department of Laboratory Medicine, Institute of Pathology, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy;
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Pasquale Persico
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (M.S.); (A.D.); (P.P.)
- IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Beatrice Casini
- Pathology Unit, Regina Elena National Cancer Institute, 00161 Rome, Italy; (M.C.); (B.C.)
| | - Matteo Fassan
- Department of Oncology, Veneto Institute of Oncology, IOV-IRCCS, 35128 Padua, Italy;
- Cytopathology Unit, Department of Medicine (DIMED), Surgical Pathology & AMP, University of Padua, 35128 Padua, Italy
| | - Vittorina Zagonel
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (V.Z.); (G.L.)
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (V.Z.); (G.L.)
| |
Collapse
|
5
|
Zappe K, Pirker C, Miedl H, Schreiber M, Heffeter P, Pfeiler G, Hacker S, Haslik W, Spiegl-Kreinecker S, Cichna-Markl M. Discrimination between 34 of 36 Possible Combinations of Three C>T SNP Genotypes in the MGMT Promoter by High Resolution Melting Analysis Coupled with Pyrosequencing Using A Single Primer Set. Int J Mol Sci 2021; 22:ijms222212527. [PMID: 34830407 PMCID: PMC8621402 DOI: 10.3390/ijms222212527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 11/12/2021] [Indexed: 11/22/2022] Open
Abstract
Due to its cost-efficiency, high resolution melting (HRM) analysis plays an important role in genotyping of candidate single nucleotide polymorphisms (SNPs). Studies indicate that HRM analysis is not only suitable for genotyping individual SNPs, but also allows genotyping of multiple SNPs in one and the same amplicon, although with limited discrimination power. By targeting the three C>T SNPs rs527559815, rs547832288, and rs16906252, located in the promoter of the O6-methylguanine-DNA methyltransferase (MGMT) gene within a distance of 45 bp, we investigated whether the discrimination power can be increased by coupling HRM analysis with pyrosequencing (PSQ). After optimizing polymerase chain reaction (PCR) conditions, PCR products subjected to HRM analysis could directly be used for PSQ. By analyzing oligodeoxynucleotide controls, representing the 36 theoretically possible variant combinations for diploid human cells (8 triple-homozygous, 12 double-homozygous, 12 double-heterozygous and 4 triple-heterozygous combinations), 34 out of the 36 variant combinations could be genotyped unambiguously by combined analysis of HRM and PSQ data, compared to 22 variant combinations by HRM analysis and 16 variant combinations by PSQ. Our approach was successfully applied to genotype stable cell lines of different origin, primary human tumor cell lines from glioma patients, and breast tissue samples.
Collapse
Affiliation(s)
- Katja Zappe
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria;
| | - Christine Pirker
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.P.); (P.H.)
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (H.M.); (M.S.)
| | - Heidi Miedl
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (H.M.); (M.S.)
- Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria
| | - Martin Schreiber
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (H.M.); (M.S.)
- Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria
| | - Petra Heffeter
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, 1090 Vienna, Austria; (C.P.); (P.H.)
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; (H.M.); (M.S.)
| | - Georg Pfeiler
- Department of Obstetrics and Gynecology, Division of Gynecology and Gynecological Oncology, Medical University of Vienna, 1090 Vienna, Austria; (G.P.); (W.H.)
| | - Stefan Hacker
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, 1090 Vienna, Austria;
- Department of Plastic, Reconstructive and Aesthetic Surgery, Landesklinikum Wiener Neustadt, 2700 Wiener Neustadt, Austria
| | - Werner Haslik
- Department of Obstetrics and Gynecology, Division of Gynecology and Gynecological Oncology, Medical University of Vienna, 1090 Vienna, Austria; (G.P.); (W.H.)
| | - Sabine Spiegl-Kreinecker
- Department of Neurosurgery, Medical Faculty, Kepler University Hospital GmbH, Johannes Kepler University Linz, 4040 Linz, Austria;
| | - Margit Cichna-Markl
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria;
- Correspondence:
| |
Collapse
|
6
|
Chen B, He A, Bi J, Sun S, Ma Y, Wang W, Guo D, Chen J, Qian Y, Shi T, Nie G, Zhao Z, Shi J, Yang H, Zhang L, Lu W. Long-range gene regulation network of the MGMT enhancer modulates glioma cell sensitivity to temozolomide. J Genet Genomics 2021; 48:946-949. [PMID: 34417124 DOI: 10.1016/j.jgg.2021.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/08/2021] [Accepted: 06/16/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Bohan Chen
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Anshun He
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jinfang Bi
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shupeng Sun
- Department of Neurosurgery, Tianjin Huanhu Hospital, School of Medicine, Nankai University, 6 Jizhao Road, Tianjin 300350, China
| | - Yiping Ma
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Wenbin Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Dianhao Guo
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jun Chen
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yuyang Qian
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Tengfei Shi
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Guohui Nie
- Department of Otolaryngology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Zhongfang Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jiandang Shi
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Hongzhen Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Lei Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| | - Wange Lu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| |
Collapse
|
7
|
Brandner S, McAleenan A, Kelly C, Spiga F, Cheng HY, Dawson S, Schmidt L, Faulkner CL, Wragg C, Jefferies S, Higgins JPT, Kurian KM. 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. Neuro Oncol 2021; 23:1457-1469. [PMID: 34467991 PMCID: PMC8408882 DOI: 10.1093/neuonc/noab105] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
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.
Collapse
Affiliation(s)
- Sebastian Brandner
- Division of Neuropathology and Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Alexandra McAleenan
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Claire Kelly
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Francesca Spiga
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Hung-Yuan Cheng
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Sarah Dawson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lena Schmidt
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Claire L Faulkner
- Bristol Genetics Laboratory, Pathology Sciences, Southmead Hospital, Bristol, UK
| | - Christopher Wragg
- Bristol Genetics Laboratory, Pathology Sciences, Southmead Hospital, Bristol, UK
| | - Sarah Jefferies
- Department of Oncology, Addenbrooke’s Hospital, Cambridge, UK
| | - Julian P T Higgins
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Kathreena M Kurian
- Bristol Medical School, Brain Tumour Research Centre, Population Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
8
|
Sim HW, McDonald KL, Lwin Z, Barnes EH, Rosenthal M, Foote MC, Koh ES, Back M, Wheeler H, Sulman EP, Buckland ME, Fisher L, Leonard R, Hall M, Ashley DM, Yip S, Simes J, Khasraw M. A randomized phase II trial of veliparib, radiotherapy and temozolomide in patients with unmethylated MGMT glioblastoma: the VERTU study. Neuro Oncol 2021; 23:1736-1749. [PMID: 33984151 PMCID: PMC8485443 DOI: 10.1093/neuonc/noab111] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Temozolomide offers minimal benefit in patients with glioblastoma with unmethylated O6-methylguanine-DNA methyltransferase (MGMT) promoter status, hence, the need for novel therapies. This study evaluated whether veliparib, a brain-penetrant poly(ADP-ribose) polymerase (PARP) inhibitor, acts synergistically with radiation and temozolomide. Methods VERTU was a multicenter 2:1 randomized phase II trial in patients with newly diagnosed glioblastoma and MGMT-unmethylated promotor status. The experimental arm consisted of veliparib and radiotherapy, followed by adjuvant veliparib and temozolomide. The standard arm consisted of concurrent temozolomide and radiotherapy, followed by adjuvant temozolomide. The primary objective was to extend the progression-free survival rate at six months (PFS-6m) in the experimental arm. Results A total of 125 participants were enrolled, with 84 in the experimental arm and 41 in the standard arm. The median age was 61 years, 70% were male, 59% had Eastern Cooperative Oncology Group (ECOG) performance status of 0, and 87% underwent macroscopic resection. PFS-6m was 46% (95% confidence interval [CI]: 36%-57%) in the experimental arm and 31% (95% CI: 18%-46%) in the standard arm. Median overall survival was 12.7 months (95% CI: 11.4-14.5 months) in the experimental arm and 12.8 months (95% CI: 9.5-15.8 months) in the standard arm. The most common grade 3-4 adverse events were thrombocytopenia and neutropenia, with no new safety signals. Conclusion The veliparib-containing regimen was feasible and well tolerated. However, there was insufficient evidence of clinical benefit in this population. Further information from correlative translational work and other trials of PARP inhibitors in glioblastoma are still awaited.
Collapse
Affiliation(s)
- Hao-Wen Sim
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, Australia.,Department of Medical Oncology, The Kinghorn Cancer Centre, Sydney, Australia.,Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, Australia
| | - Kerrie L McDonald
- Cure Brain Cancer Neuro-Oncology Lab, University of New South Wales, Sydney, Australia
| | - Zarnie Lwin
- School of Medicine, University of Queensland, Brisbane, Australia.,Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | | | - Mark Rosenthal
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Department of Medical Oncology, Royal Melbourne Hospital, Melbourne, Australia
| | - Matthew C Foote
- School of Medicine, University of Queensland, Brisbane, Australia.,Department of Radiation Oncology, Princess Alexandra Hospital, Brisbane, Australia
| | - Eng-Siew Koh
- South Western Sydney Clinical School, University of New South Wales, Sydney, Australia.,Ingham Institute for Applied Medical Research, Sydney, Australia.,Department of Radiation Oncology, Liverpool Hospital, Sydney, Australia
| | - Michael Back
- Department of Radiation Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Helen Wheeler
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Erik P Sulman
- Department of Radiation Oncology, NYU Grossman School of Medicine and Brain and Spine Tumors, New York, USA.,Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, USA
| | - Michael E Buckland
- Neuropathology Department, Royal Prince Alfred Hospital, Sydney, Australia.,Brain and Mind Centre, University of Sydney, Sydney, Australia
| | - Lauren Fisher
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Robyn Leonard
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Merryn Hall
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - David M Ashley
- Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Sonia Yip
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia.,Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, Australia
| | - Mustafa Khasraw
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia.,Duke University School of Medicine, Duke University, Durham, NC, USA
| |
Collapse
|
9
|
McAleenan A, Kelly C, Spiga F, Kernohan A, Cheng HY, Dawson S, Schmidt L, Robinson T, Brandner S, Faulkner CL, Wragg C, Jefferies S, Howell A, Vale L, Higgins JPT, Kurian KM. Prognostic value of test(s) for O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation for predicting overall survival in people with glioblastoma treated with temozolomide. Cochrane Database Syst Rev 2021; 3:CD013316. [PMID: 33710615 PMCID: PMC8078495 DOI: 10.1002/14651858.cd013316.pub2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Glioblastoma is an aggressive form of brain cancer. Approximately five in 100 people with glioblastoma survive for five years past diagnosis. Glioblastomas that have a particular modification to their DNA (called methylation) in a particular region (the O6-methylguanine-DNA methyltransferase (MGMT) promoter) respond better to treatment with chemotherapy using a drug called temozolomide. OBJECTIVES To determine which method for assessing MGMT methylation status best predicts overall survival in people diagnosed with glioblastoma who are treated with temozolomide. SEARCH METHODS We searched MEDLINE, Embase, BIOSIS, Web of Science Conference Proceedings Citation Index to December 2018, and examined reference lists. For economic evaluation studies, we additionally searched NHS Economic Evaluation Database (EED) up to December 2014. SELECTION CRITERIA Eligible studies were longitudinal (cohort) studies of adults with diagnosed glioblastoma treated with temozolomide with/without radiotherapy/surgery. Studies had to have related MGMT status in tumour tissue (assessed by one or more method) with overall survival and presented results as hazard ratios or with sufficient information (e.g. Kaplan-Meier curves) for us to estimate hazard ratios. We focused mainly on studies comparing two or more methods, and listed brief details of articles that examined a single method of measuring MGMT promoter methylation. We also sought economic evaluations conducted alongside trials, modelling studies and cost analysis. DATA COLLECTION AND ANALYSIS Two review authors independently undertook all steps of the identification and data extraction process for multiple-method studies. We assessed risk of bias and applicability using our own modified and extended version of the QUality In Prognosis Studies (QUIPS) tool. We compared different techniques, exact promoter regions (5'-cytosine-phosphate-guanine-3' (CpG) sites) and thresholds for interpretation within studies by examining hazard ratios. We performed meta-analyses for comparisons of the three most commonly examined methods (immunohistochemistry (IHC), methylation-specific polymerase chain reaction (MSP) and pyrosequencing (PSQ)), with ratios of hazard ratios (RHR), using an imputed value of the correlation between results based on the same individuals. MAIN RESULTS We included 32 independent cohorts involving 3474 people that compared two or more methods. We found evidence that MSP (CpG sites 76 to 80 and 84 to 87) is more prognostic than IHC for MGMT protein at varying thresholds (RHR 1.31, 95% confidence interval (CI) 1.01 to 1.71). We also found evidence that PSQ is more prognostic than IHC for MGMT protein at various thresholds (RHR 1.36, 95% CI 1.01 to 1.84). The data suggest that PSQ (mainly at CpG sites 74 to 78, using various thresholds) is slightly more prognostic than MSP at sites 76 to 80 and 84 to 87 (RHR 1.14, 95% CI 0.87 to 1.48). Many variants of PSQ have been compared, although we did not see any strong and consistent messages from the results. Targeting multiple CpG sites is likely to be more prognostic than targeting just one. In addition, we identified and summarised 190 articles describing a single method for measuring MGMT promoter methylation status. AUTHORS' CONCLUSIONS PSQ and MSP appear more prognostic for overall survival than IHC. Strong evidence is not available to draw conclusions with confidence about the best CpG sites or thresholds for quantitative methods. MSP has been studied mainly for CpG sites 76 to 80 and 84 to 87 and PSQ at CpG sites ranging from 72 to 95. A threshold of 9% for CpG sites 74 to 78 performed better than higher thresholds of 28% or 29% in two of three good-quality studies making such comparisons.
Collapse
Affiliation(s)
- Alexandra McAleenan
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Claire Kelly
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Francesca Spiga
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ashleigh Kernohan
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Hung-Yuan Cheng
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Sarah Dawson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Applied Research Collaboration West (ARC West) , University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Lena Schmidt
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tomos Robinson
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Claire L Faulkner
- Bristol Genetics Laboratory, Pathology Sciences, Southmead Hospital, Bristol, UK
| | - Christopher Wragg
- Bristol Genetics Laboratory, Pathology Sciences, Southmead Hospital, Bristol, UK
| | - Sarah Jefferies
- Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| | - Amy Howell
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Luke Vale
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Julian P T Higgins
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Applied Research Collaboration West (ARC West) , University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Kathreena M Kurian
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol Medical School: Brain Tumour Research Centre, Public Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
10
|
Farrell C, Shi W, Bodman A, Olson JJ. Congress of neurological surgeons systematic review and evidence-based guidelines update on the role of emerging developments in the management of newly diagnosed glioblastoma. J Neurooncol 2020; 150:269-359. [PMID: 33215345 DOI: 10.1007/s11060-020-03607-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/23/2020] [Indexed: 12/12/2022]
Abstract
TARGET POPULATION These recommendations apply to adult patients with newly diagnosed or suspected glioblastoma. IMAGING Question What imaging modalities are in development that may be able to provide improvements in diagnosis, and therapeutic guidance for individuals with newly diagnosed glioblastoma? RECOMMENDATION Level III: It is suggested that techniques utilizing magnetic resonance imaging for diffusion weighted imaging, and to measure cerebral blood and magnetic spectroscopic resonance imaging of N-acetyl aspartate, choline and the choline to N-acetyl aspartate index to assist in diagnosis and treatment planning in patients with newly diagnosed or suspected glioblastoma. SURGERY Question What new surgical techniques can be used to provide improved tumor definition and resectability to yield better tumor control and prognosis for individuals with newly diagnosed glioblastoma? RECOMMENDATIONS Level II: The use of 5-aminolevulinic acid is recommended to improve extent of tumor resection in patients with newly diagnosed glioblastoma. Level II: The use of 5-aminolevulinic acid is recommended to improve median survival and 2 year survival in newly diagnosed glioblastoma patients with clinical characteristics suggesting poor prognosis. Level III: It is suggested that, when available, patients be enrolled in properly designed clinical trials assessing the value of diffusion tensor imaging in improving the safety of patients with newly diagnosed glioblastoma undergoing surgery. NEUROPATHOLOGY Question What new pathology techniques and measurement of biomarkers in tumor tissue can be used to provide improved diagnostic ability, and determination of therapeutic responsiveness and prognosis for patients with newly diagnosed glioblastomas? RECOMMENDATIONS Level II: Assessment of tumor MGMT promoter methylation status is recommended as a significant predictor of a longer progression free survival and overall survival in patients with newly diagnosed with glioblastoma. Level II: Measurement of tumor expression of neuron-glia-2, neurofilament protein, glutamine synthetase and phosphorylated STAT3 is recommended as a predictor of overall survival in patients with newly diagnosed with glioblastoma. Level III: Assessment of tumor IDH1 mutation status is suggested as a predictor of longer progression free survival and overall survival in patients with newly diagnosed with glioblastoma. Level III: Evaluation of tumor expression of Phosphorylated Mitogen-Activated Protein Kinase protein, EGFR protein, and Insulin-like Growth Factor-Binding Protein-3 is suggested as a predictor of overall survival in patients with newly diagnosed with glioblastoma. RADIATION Question What radiation therapy techniques are in development that may be used to provide improved tumor control and prognosis for individuals with newly diagnosed glioblastomas? RECOMMENDATIONS Level III: It is suggested that patients with newly diagnosed glioblastoma undergo pretreatment radio-labeled amino acid tracer positron emission tomography to assess areas at risk for tumor recurrence to assist in radiation treatment planning. Level III: It is suggested that, when available, patients be with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of radiation dose escalation, altered fractionation, or new radiation delivery techniques. CHEMOTHERAPY Question What emerging chemotherapeutic agents or techniques are available to provide better tumor control and prognosis for patients with newly diagnosed glioblastomas? RECOMMENDATION Level III: As no emerging chemotherapeutic agents or techniques were identified in this review that improved tumor control and prognosis it is suggested that, when available, patients with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of chemotherapy. MOLECULAR AND TARGETED THERAPY Question What new targeted therapy agents are available to provide better tumor control and prognosis for individuals with newly diagnosed glioblastomas? RECOMMENDATION Level III: As no new molecular and targeted therapies have clearly provided better tumor control and prognosis it is suggested that, when available, patients with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of molecular and targeted therapies IMMUNOTHERAPY: Question What emerging immunotherapeutic agents or techniques are available to provide better tumor control and prognosis for patients with newly diagnosed glioblastomas? RECOMMENDATION Level III: As no immunotherapeutic agents have clearly provided better tumor control and prognosis it is suggested that, when available, patients with newly diagnosed glioblastomas be enrolled in properly designed clinical trials of immunologically-based therapies. NOVEL THERAPIES Question What novel therapies or techniques are in development to provide better tumor control and prognosis for individuals with newly diagnosed glioblastomas? RECOMMENDATIONS Level II: The use of tumor-treating fields is recommended for patients with newly diagnosed glioblastoma who have undergone surgical debulking and completed concurrent chemoradiation without progression of disease at the time of tumor-treating field therapy initiation. Level II: It is suggested that, when available, enrollment in properly designed studies of vector containing herpes simplex thymidine kinase gene and prodrug therapies be considered in patients with newly diagnosed glioblastoma.
Collapse
Affiliation(s)
- Christopher Farrell
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Wenyin Shi
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
11
|
Biau J, Thivat E, Chautard E, Stefan D, Boone M, Chauffert B, Bourgne C, Richard D, Molnar I, Levesque S, Bellini R, Kwiatkowski F, Karayan-Tapon L, Verrelle P, Godfraind C, Durando X. Phase 1 trial of ralimetinib (LY2228820) with radiotherapy plus concomitant temozolomide in the treatment of newly diagnosed glioblastoma. Radiother Oncol 2020; 154:227-234. [PMID: 32976869 DOI: 10.1016/j.radonc.2020.09.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND AND PURPOSE This phase 1 trial aimed to determine the maximum tolerated dose (MTD; primary objective) of a p38-MAPK inhibitor, ralimetinib, with radiotherapy (RT) and chemotherapy (TMZ), in the treatment of newly diagnosed glioblastoma (GBM) patients. MATERIALS AND METHODS The study was designed as an open-label dose-escalation study driven by a Tite-CRM design and followed by an expansion cohort. Ralimetinib was administered orally every 12 h, 7 days a week, for 2 cycles of 2 weeks at a dose of 100, 200 or 300 mg/12 h. Patients received ralimetinib added to standard concurrent RT (60 Gy in 30 fractions) with TMZ (75 mg/m2/day) and 6 cycles of adjuvant TMZ (150-200 mg/m2 on days 1-5 every 28 days). RESULTS The MTD of ralimetinib was 100 mg/12 h with chemoradiotherapy. The three patients treated at 200 mg/12 h presented a dose-limiting toxicity: one patient had a grade 3 face edema, and two patients had a grade 3 rash and grade 3 hepatic cytolysis (66%). Of the 18 enrolled patients, 15 received the MTD of ralimetinib. At the MTD, the grade ≥ 3 adverse events during concomitant chemoradiotherapy were hepatic cytolysis (2/15 patients), dermatitis/rash (1/15), lymphopenia (1/15) and nausea/vomiting (1/15). No interaction of TMZ and ralimetinib when administrated concomitantly has been observed. Inhibition of pMAPKAP-K2 (-54%) was observed in peripheral blood mononuclear cells. CONCLUSION This phase 1 trial is the first trial to study the combination of a p38-MAPK inhibitor, ralimetinib, with radiotherapy (RT) and chemotherapy (TMZ), in the treatment of newly diagnosed glioblastoma (GBM) patients. The MTD of ralimetinib was 100 mg/12 h. The most frequent dose-limiting toxicities were hepatic cytolysis and rash.
Collapse
Affiliation(s)
- J Biau
- Radiation Department, Centre Jean Perrin, Clermont-Ferrand, France; University of Clermont Auvergne, UFR Médecine, Clermont-Ferrand, France; INSERM U1240 IMoST, University of Clermont Auvergne, Clermont-Ferrand, France; Centre d'Investigation Clinique UMR 501, Clermont-Ferrand, France.
| | - E Thivat
- INSERM U1240 IMoST, University of Clermont Auvergne, Clermont-Ferrand, France; Centre d'Investigation Clinique UMR 501, Clermont-Ferrand, France; Department of Clinical Research, Délégation Recherche Clinique et Innovation, Centre Jean Perrin, Clermont-Ferrand, France
| | - E Chautard
- INSERM U1240 IMoST, University of Clermont Auvergne, Clermont-Ferrand, France; Pathology Department, Centre Jean Perrin, Clermont-Ferrand, France
| | - D Stefan
- Radiation Oncology Department, Centre François Baclesse, Caen, France
| | - M Boone
- Department of Medical oncology, CHU Amiens, France
| | - B Chauffert
- Department of Medical oncology, CHU Amiens, France
| | - C Bourgne
- Department of Biologic hematology, CHU Estaing, Clermont-Ferrand Cedex 1, France
| | - D Richard
- CHU Clermont-Ferrand, University of Clermont-Auvergne, Medical Pharmacology Department, UMR INSERM, Clermont-Ferrand, France
| | - I Molnar
- INSERM U1240 IMoST, University of Clermont Auvergne, Clermont-Ferrand, France; Centre d'Investigation Clinique UMR 501, Clermont-Ferrand, France; Department of Clinical Research, Délégation Recherche Clinique et Innovation, Centre Jean Perrin, Clermont-Ferrand, France
| | - S Levesque
- INSERM U1240 IMoST, University of Clermont Auvergne, Clermont-Ferrand, France; Centre d'Investigation Clinique UMR 501, Clermont-Ferrand, France; Department of Clinical Research, Délégation Recherche Clinique et Innovation, Centre Jean Perrin, Clermont-Ferrand, France
| | - R Bellini
- Radiodiagnostic Department, Centre Jean-Perrin, Clermont-Ferrand, France
| | - F Kwiatkowski
- INSERM U1240 IMoST, University of Clermont Auvergne, Clermont-Ferrand, France; Centre d'Investigation Clinique UMR 501, Clermont-Ferrand, France; Department of Clinical Research, Délégation Recherche Clinique et Innovation, Centre Jean Perrin, Clermont-Ferrand, France
| | - L Karayan-Tapon
- University of Poitiers, INSERMU1084, CHU de Poitiers, Department of Cancer Biology, France
| | - P Verrelle
- Radiation Department, Centre Jean Perrin, Clermont-Ferrand, France; University of Clermont Auvergne, UFR Médecine, Clermont-Ferrand, France; Department of Radiation Oncology, Institut Curie, Paris, France
| | - C Godfraind
- Department of Pathological Anatomy, CHU de Clermont-Ferrand, France
| | - X Durando
- INSERM U1240 IMoST, University of Clermont Auvergne, Clermont-Ferrand, France; Centre d'Investigation Clinique UMR 501, Clermont-Ferrand, France; Department of Clinical Research, Délégation Recherche Clinique et Innovation, Centre Jean Perrin, Clermont-Ferrand, France; Oncology Department, Centre Jean Perrin, Clermont-Ferrand, France; University of Clermont Auvergne, UFR Médecine, Clermont-Ferrand, France
| |
Collapse
|
12
|
Ma L, Liang B, Yang Y, Chen L, Liu Q, Zhang A. hOGG1 promoter methylation, hOGG1 genetic variants and their interactions for risk of coal-borne arsenicosis: A case-control study. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 75:103330. [PMID: 32004920 DOI: 10.1016/j.etap.2020.103330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 06/10/2023]
Abstract
To identify the effect of hOGG1 methylation, Ser326Cys polymorphism and their interactions on the risk of coal-borne arsenicosis, 113 coal-borne arsenicosis subjects and 55 reference subjects were recruited. Urinary arsenic contents were analyzed with ICP-MS. hOGG1 methylation and Ser326Cys polymorphism was measured by mehtylation-specific PCR and restriction fragment length polymorphism PCR in PBLCs, respectively. The results showed that the prevalence of methylated hOGG1 and variation genotype (326 Ser/Cys & 326 Cys/Cys) were increased with raised levels of urinary arsenic in arsenicosis subjects. Increased prevalence of methylated hOGG1 and variation genotype were associated with raised risk of arsenicosis. Moreover, the results revealed that variant genotype might increase the susceptibility to hOGG1 methylation. The interactions of methylated hOGG1 and variation genotype were also found to contribute to increased risk of arsenicosis. Taken together, hOGG1 hypermethylation, hOGG1 variants and their interactions might be potential biomarkers for evaluating risk of coal-borne arsenicosis.
Collapse
Affiliation(s)
- Lu Ma
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China.
| | - Bing Liang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China.
| | - Yuan Yang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China.
| | - Liyuan Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China.
| | - Qizhan Liu
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, PR China.
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China.
| |
Collapse
|
13
|
Jue TR, Olafson LR, Siddell AH, Rapkins RW, Ng B, Yin JXM, Lu VM, Chung SA, Whittaker SP, Davies M, Fairhall JM, Hovey EJ, McDonald KL. A case study of a long-term glioblastoma survivor with unmethylated MGMT and hypermutated genotype. Cold Spring Harb Mol Case Stud 2019; 5:a003251. [PMID: 31160353 PMCID: PMC6549560 DOI: 10.1101/mcs.a003251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 03/25/2019] [Indexed: 12/19/2022] Open
Abstract
Effective treatments that extend survival of malignant brain tumor glioblastoma (GBM) have not changed in more than a decade; however, there exists a minority patient group (<5%) whose survival is longer than 3 yr. We herein present a case report of a long-term surviving 51-yr-old female diagnosed with a MGMT unmethylated GBM. The patient was progression-free for 23 mo. Fresh primary and recurrent tumor samples were collected and processed for patient-derived model development. Whole-genome sequencing (WGS) was performed concurrently with additional standard of care diagnostics. WGS revealed a hypermutated genotype in the germline tissue and in both the primary and recurrent tumor samples. Specific to the matched tumors, an average of 30 cancer driver genes were mutated. Noteworthy was the identification of a nonsynonymous mutation in the POLE gene. As a possible instigator of the hypermutational genotype observed in the tumors, we identified nonsynonymous germline mutations within the mismatch repair genes, MLH1 and PMS2 Mutations within these genes are often indicative of the pan-cancer phenotype known as Lynch syndrome; however, their pathogenicity remains unreported. We performed a drug screen of 165 compounds, which identified one compound, YM155, an experimental survivin inhibitor, that showed effectivity to the patient-derived cell lines of both tumors. Treatment selection based on a patient's genome to individualize treatment for GBM patients could potentially be useful in the clinic. This is a promising avenue for further translational research, with larger databases and integrated platforms to increase the efficiency of analyzing and interpreting the individual genomic data of GBM.
Collapse
Affiliation(s)
- Toni Rose Jue
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Lauren R Olafson
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Anna H Siddell
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Robert W Rapkins
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Benedict Ng
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Julia X M Yin
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Victor M Lu
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Sylvia A Chung
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Shane P Whittaker
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Matthew Davies
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Jacob M Fairhall
- Neurospine Clinic, Prince of Wales Hospital, Randwick, New South Wales 2031, Australia
- University of New South Wales, Sydney, New South Wales 2031, Australia
| | - Elizabeth J Hovey
- University of New South Wales, Sydney, New South Wales 2031, Australia
- Department of Medical Oncology, Nelune Comprehensive Cancer Centre, Prince of Wales, Hospital, Randwick, New South Wales 2031, Australia
| | - Kerrie L McDonald
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
14
|
Sánchez‐Siles M, Aliaga‐Sánchez A, Medina S, Adoamnei E, Fernández‐Ruiz JA, Pelegrín‐Hernández JP, Corno‐Caparrós A, Rosa‐Salazar V, Camacho‐Alonso F. Genotyping of the C>T allele of rs16906252, predictor of O16‐methylguanine‐DNA methyltransferase (MGMT) promoter methylation status, in erosive atrophic lesions of oral lichen planus. Int J Dermatol 2019; 58:1078-1082. [DOI: 10.1111/ijd.14473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 02/18/2019] [Accepted: 04/11/2019] [Indexed: 12/11/2022]
Affiliation(s)
| | - Alfonso Aliaga‐Sánchez
- Oral and Maxillofacial Surgery Reina Sofia Hospital Murcia Spain
- Department of Oral Surgery University of Murcia Murcia Spain
| | - Sonia Medina
- Department of Food Science and Technology CEBAS‐CSIC Murcia Spain
| | - Evdochia Adoamnei
- Department of Public Health Sciences University of Murcia Murcia Spain
| | | | | | | | - Vladimir Rosa‐Salazar
- Tromboembolic Disease Unit/Short Stay Unit Virgen de la Arrixaca University Hospital Murcia Spain
| | | |
Collapse
|
15
|
Ibudilast sensitizes glioblastoma to temozolomide by targeting Macrophage Migration Inhibitory Factor (MIF). Sci Rep 2019; 9:2905. [PMID: 30814573 PMCID: PMC6393433 DOI: 10.1038/s41598-019-39427-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/04/2018] [Indexed: 02/06/2023] Open
Abstract
Recurrence in patients with glioblastoma (GBM) is inevitable resulting in short survival times, even in patients with O-6-Methylguanine-DNA Methyltransferase (MGMT) methylation. Other pathways must be activated to escape from temozolomide (TMZ) treatment, however acquired resistance mechanisms to TMZ are not well understood. Herein, frozen tumors from 36 MGMT methylated patients grouped according to overall survival were extracted and proteins were profiled using surface-enhanced laser desorption/ionization (SELDI) with time-of flight (TOF) proteomics to identify low molecular weight proteins that associated with poor survival outcomes. Overexpression of macrophage migration inhibitory factor (MIF) was identified in human GBM specimens that were MGMT methylated but showed poor survival. This correlation was confirmed in an independent cohort of human GBM. MIF overexpression has been reported in several cancer types, including GBM. We repurposed ibudilast, a specific MIF inhibitor, and treated patient derived cell lines. Ibudilast showed modest anti-proliferative activity however, when combined with TMZ, significant synergism was observed, resulting in cell cycle arrest and apoptosis. In vivo, combined ibudilast and TMZ treatment of a patient derived xenograft (PDX) model resulted in significantly longer overall survival. Our findings have significant clinical implications for people with GBM. Since clinical trials involving ibudilast have shown no adverse side effects and the drug readily penetrates the blood brain barrier, treatment of GBM with this combination is clinically achievable.
Collapse
|
16
|
Huang L, Xu W, Dai L, Yan D, Zhang S, Shi X. An intronic genetic variation of MGMT affects enhancer activity and is associated with glioma susceptibility. Cancer Manag Res 2018; 10:3995-4003. [PMID: 30310321 PMCID: PMC6166748 DOI: 10.2147/cmar.s176622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose O6-methylguanine-DNA methyltransferase (MGMT) plays a crucial role in repairing damaged DNA caused by alkylating agents. A number of cancer susceptibility loci have been recognized as enhancer variants. This study aimed to explore the significance of enhancer variants of MGMT in glioma susceptibility. Patients and methods A retrospective case-control study consisting of 150 glioma patients and 327 controls was conducted to test whether enhancer variants of MGMT are associated with glioma susceptibility. Genotypes were determined by Sequenom MassARRAY technology. Associations were estimated by logistic regression. Biochemical assays were used to examine the function of glioma susceptibility locus. Results We found that the A allele of rs10764901, an intronic variant of MGMT, was associated with a significantly decreased risk of glioma. The rs10764901 AA genotype carriers had an OR of 0.49 (95% CI, 0.24-0.98; P=0.045) compared with the rs10764901 GG genotype. When the rs10764901 AG and AA genotypes were pooled for analysis, a significantly decreased risk of glioma was also found (OR, 0.63; 95% CI, 0.43-0.93; P=0.021). Functional analyses showed that the rs10764901 A allele drove a lower luciferase expression and had higher transcription factor binding affinity than the G allele. Conclusion An enhancer variant of MGMT rs10764901 affects the regulatory activity of enhancer by altering the binding affinity of transcription factors and is associated with glioma susceptibility.
Collapse
Affiliation(s)
- Liming Huang
- The First Department of Chemotherapy, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China,
| | - Wenshen Xu
- Department of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Lian Dai
- Department of Medicine, The Third Affiliated People's Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Danfang Yan
- Department of Radiation Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shu Zhang
- The First Department of Chemotherapy, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China,
| | - Xi Shi
- The First Department of Chemotherapy, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China,
| |
Collapse
|
17
|
Hombach-Klonisch S, Mehrpour M, Shojaei S, Harlos C, Pitz M, Hamai A, Siemianowicz K, Likus W, Wiechec E, Toyota BD, Hoshyar R, Seyfoori A, Sepehri Z, Ande SR, Khadem F, Akbari M, Gorman AM, Samali A, Klonisch T, Ghavami S. Glioblastoma and chemoresistance to alkylating agents: Involvement of apoptosis, autophagy, and unfolded protein response. Pharmacol Ther 2018; 184:13-41. [DOI: 10.1016/j.pharmthera.2017.10.017] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
18
|
Cheng G, Xin Q, Li K, Sun C, Lin Z, Luan Y, Wang J, Qi T, Chen Y, Xu D, Kong F. The Association between O(6)-Methylguanine DNA Methyltransferase (MGMT) rs11016879 and rs7069143 Polymorphisms and Susceptibility to Еsophageal Аdenocarcinoma in a Han Chinese Population. RUSS J GENET+ 2018. [DOI: 10.1134/s1022795418040063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Abstract
Malignant glioma is a devastating disease affecting both adults and children with limited treatment strategies. Pre-clinical animal studies are critical to the development and planning of novel treatment designs for human clinical trials. Topoisomerases has been a target of interest in the treatment of high grade gliomas, such as glioblastoma, in the past years. Here we assess pre-clinical glioma literature with the aim to identify predictive variables that favour treatment outcomes from topoisomerase inhibition. Data was extracted from 90 experimental comparisons, this was divided based on available survival (n = 61) and tumor volume (n = 29) data. The meta-analysis revealed that the overall effect of topoisomerase inhibition prolonged survival by a factor of 1.33 (95% CI: 1.23–1.43) and reduced tumor growth by a factor of 3.21 (95% CI: 1.99–5.88), with considerable between-study heterogeneity. Multivariable meta-regression identified glioma model, type of control, route of drug administration and drug of choice to be predictive of improved survival outcome. Publication bias assessment by contour-enhanced funnel plots, Egger’s regression test and trim and fill analysis showed evidence of publication bias in all studies. This study identified multiple study design factors that should be taken into consideration to improve the translation of pre-clinical investigation of topoisomerase inhibition into clinical use.
Collapse
|
20
|
de Boni L, Gasparoni G, Haubenreich C, Tierling S, Schmitt I, Peitz M, Koch P, Walter J, Wüllner U, Brüstle O. DNA methylation alterations in iPSC- and hESC-derived neurons: potential implications for neurological disease modeling. Clin Epigenetics 2018; 10:13. [PMID: 29422978 PMCID: PMC5789607 DOI: 10.1186/s13148-018-0440-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/04/2018] [Indexed: 12/21/2022] Open
Abstract
Background Genetic predisposition and epigenetic alterations are both considered to contribute to sporadic neurodegenerative diseases (NDDs) such as Parkinson's disease (PD). Since cell reprogramming and the generation of induced pluripotent stem cells (iPSCs) are themselves associated with major epigenetic remodeling, it remains unclear to what extent iPSC-derived neurons lend themselves to model epigenetic disease-associated changes. A key question to be addressed in this context is whether iPSC-derived neurons exhibit epigenetic signatures typically observed in neurons derived from non-reprogrammed human embryonic stem cells (hESCs). Results Here, we compare mature neurons derived from hESC and isogenic human iPSC generated from hESC-derived neural stem cells. Genome-wide 450 K-based DNA methylation and HT12v4 gene array expression analyses were complemented by a deep analysis of selected genes known to be involved in NDD. Our studies show that DNA methylation and gene expression patterns of isogenic hESC- and iPSC-derived neurons are markedly preserved on a genome-wide and single gene level. Conclusions Overall, iPSC-derived neurons exhibit similar DNA methylation patterns compared to isogenic hESC-derived neurons. Further studies will be required to explore whether the epigenetic patterns observed in iPSC-derived neurons correspond to those detectable in native brain neurons.
Collapse
Affiliation(s)
- Laura de Boni
- Department of Neurology, University Hospital of Bonn, Bonn, Germany
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Gilles Gasparoni
- Institute for Genetics/Epigenetics, FR8.3 Life Sciences, Saarland University, Saarbrücken, Germany
| | - Carolin Haubenreich
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Sascha Tierling
- Institute for Genetics/Epigenetics, FR8.3 Life Sciences, Saarland University, Saarbrücken, Germany
| | - Ina Schmitt
- Department of Neurology, University Hospital of Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany
| | - Philipp Koch
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Jörn Walter
- Institute for Genetics/Epigenetics, FR8.3 Life Sciences, Saarland University, Saarbrücken, Germany
| | - Ullrich Wüllner
- Department of Neurology, University Hospital of Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, Life & Brain Center, University of Bonn, Bonn, Germany
| |
Collapse
|
21
|
Biau J, Chautard E, De Schlichting E, Dupic G, Pereira B, Fogli A, Müller-Barthélémy M, Dalloz P, Khalil T, Dillies AF, Durando X, Godfraind C, Verrelle P. Radiotherapy plus temozolomide in elderly patients with glioblastoma: a "real-life" report. Radiat Oncol 2017; 12:197. [PMID: 29212499 PMCID: PMC5719937 DOI: 10.1186/s13014-017-0929-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/21/2017] [Indexed: 11/22/2022] Open
Abstract
Background The optimization of the management for elderly glioblastoma patients is crucial given the demographics of aging in many countries. We report the outcomes for a “real-life” patient cohort (i.e. unselected) comprising consecutive glioblastoma patients aged 70 years or more, treated with different radiotherapy +/− temozolomide regimens. Methods From 2003 to 2016, 104 patients ≥ 70 years of age, consecutively treated by radiotherapy for glioblastoma, were included in this study. All patients were diagnosed with IDH-wild type glioblastoma according to pathological criteria. Results Our patient cohort comprised 51 female patients (49%) and 53 male. The median cohort age was 75 years (70–88), and the median Karnofsky performance status (KPS) was 70 (30–100). Five (5%) patients underwent macroscopic complete resection, 9 (9%) had partial resection, and 90 (86%), a stereotactic biopsy. The MGMT promoter was methylated in 33/73 cases (45%). Fifty-two (50%), 38 (36%), and 14 (14%) patients were categorized with RPA scores of III, IV, and I-II. Thirty-three (32%) patients received normofractionated radiotherapy (60 Gy, 30 sessions) with temozolomide (Stupp), 37 (35%) received hypofractionated radiotherapy (median dose 40 Gy, 15 sessions) with temozolomide (HFRT + TMZ), and 34 (33%) HFRT alone. Patients receiving only HFRT were significantly older, with lower KPSs. The median overall survival (OS; all patients) was 5.2 months. OS rates at 12, 18, and 24 months, were 19%, 12%, and 5%, respectively, with no statistical differences between patients receiving Stupp or HFRT + TMZ (P = 0.22). In contrast, patients receiving HFRT alone manifested a significantly shorter survival time (3.9 months vs. 5.9 months, P = 0.018). In multivariate analyses, the prognostic factors for OS were: i) the type of surgery (HR: 0.47 [0.26–0.86], P = 0.014), ii) RPA class (HR: 2.15 [1.17–3.95], P = 0.014), and iii) temozolomide use irrespective of radiotherapy schedule (HR: 0.54 [0.33–0.88], P < 0.02). MGMT promoter methylation was neither a prognostic nor a predictive factor. Conclusions These outcomes agree with the literature in terms of optimal surgery and the use of HFRT as a standard treatment for elderly GBM patients. Our study emphasizes the potential benefit of using temozolomide with radiotherapy in a real-life cohort of elderly GBM patients, irrespective of their MGMT status.
Collapse
Affiliation(s)
- J Biau
- Radiotherapy Department, Université Clermont Auvergne, Centre Jean Perrin, 63011, Clermont-Ferrand, France. .,Université Clermont Auvergne, INSERM, U1240 IMoST, F-63000, Clermont Ferrand, France.
| | - E Chautard
- Radiotherapy Department, Université Clermont Auvergne, Centre Jean Perrin, 63011, Clermont-Ferrand, France.,Université Clermont Auvergne, INSERM, U1240 IMoST, F-63000, Clermont Ferrand, France
| | - E De Schlichting
- Neurosurgery Department, Clermont-Ferrand Hospital, 63003, Clermont-Ferrand, France
| | - G Dupic
- Radiotherapy Department, Université Clermont Auvergne, Centre Jean Perrin, 63011, Clermont-Ferrand, France
| | - B Pereira
- Biostatistics Department, DRCI, Clermont-Ferrand Hospital, 63003, Clermont-Ferrand, France
| | - A Fogli
- Université Clermont Auvergne, CNRS UMR 6293, INSERM U1103, GReD Laboratory, 63000, Clermont-Ferrand, France
| | - M Müller-Barthélémy
- Université Clermont Auvergne, INSERM, U1240 IMoST, F-63000, Clermont Ferrand, France
| | - P Dalloz
- Oncology Department, Université Clermont Auvergne, Centre Jean Perrin, 63011, Clermont-Ferrand, France
| | - T Khalil
- Neurosurgery Department, Clermont-Ferrand Hospital, 63003, Clermont-Ferrand, France
| | - A F Dillies
- Oncology Department, Université Clermont Auvergne, Centre Jean Perrin, 63011, Clermont-Ferrand, France
| | - X Durando
- Université Clermont Auvergne, INSERM, U1240 IMoST, F-63000, Clermont Ferrand, France.,Oncology Department, Université Clermont Auvergne, Centre Jean Perrin, 63011, Clermont-Ferrand, France
| | - C Godfraind
- Université Clermont Auvergne, INSERM, U1240 IMoST, F-63000, Clermont Ferrand, France.,Anatomopathology Department, Clermont-Ferrand Hospital, 63003, Clermont-Ferrand, France
| | - P Verrelle
- Radiotherapy Department, Université Clermont Auvergne, Centre Jean Perrin, 63011, Clermont-Ferrand, France.,Radiation Oncology Department, Institut Curie, 75248, Paris, France
| |
Collapse
|
22
|
D'Souza W, Saranath D. OMICS, Oral Cancer Molecular Landscapes, and Clinical Practice. ACTA ACUST UNITED AC 2017; 21:689-703. [DOI: 10.1089/omi.2017.0146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Wendy D'Souza
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be) University, Mumbai, India
| | - Dhananjaya Saranath
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be) University, Mumbai, India
| |
Collapse
|
23
|
Hsu CY, Ho HL, Lin SC, Ho TDH, Ho DMT. The MGMT promoter single-nucleotide polymorphism rs1625649 had prognostic impact on patients with MGMT methylated glioblastoma. PLoS One 2017; 12:e0186430. [PMID: 29036186 PMCID: PMC5643071 DOI: 10.1371/journal.pone.0186430] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/29/2017] [Indexed: 01/02/2023] Open
Abstract
Promoter methylation is the most significant mechanism to regulate O6-methylguanine-DNA-methyltransferase (MGMT) expression. Single-nucleotide polymorphisms (SNPs) in the MGMT promoter region may also play a role. The aim of this study was to evaluate the clinical significance of SNPs in the MGMT promoter region of glioblastoma. Genomic DNAs from 118 glioblastomas were collected for polymerase chain reaction (PCR) amplification. Sanger sequencing was used to sequence the MGMT promoter region to detect SNPs. The results were correlated with MGMT status and patient survival. Rs1625649 was the only polymorphic SNP located at the MGMT promoter region in 37.5% of glioblastomas. Homozygous rs1625649 (AA genotype) was correlated with a higher MGMT methylation level and a lower protein expression, but the result was not statistically significant. In patients with MGMT methylated glioblastoma, cases with homozygous rs1625649 (AA genotype) were significantly associated with a lack of MGMT protein expression and a better progression-free survival (PFS) than the cases with wild type rs1625649 (CC genotype) or heterozygous rs1625649 (CA genotype). The survival impact was significant in multivariate analyses. In conclusion, the MGMT promoter homozygous rs1625649 (AA genotype) was found to correlate with a better PFS in patients with MGMT methylated glioblastoma.
Collapse
Affiliation(s)
- Chih-Yi Hsu
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hsiang-Ling Ho
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Chieh Lin
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tiffany Dai-Hwa Ho
- Department of Computer Science and Department of Statistics, Duke University, Durham, United States of America
| | - Donald Ming-Tak Ho
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Pathology and Laboratory Medicine, Cheng Hsin General Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
24
|
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
|
25
|
Meng W, Jiang Y, Ma J. Is the prognostic significance of O6-methylguanine- DNA methyltransferase promoter methylation equally important in glioblastomas of patients from different continents? A systematic review with meta-analysis. Cancer Manag Res 2017; 9:411-425. [PMID: 29033608 PMCID: PMC5614747 DOI: 10.2147/cmar.s140447] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND O6-methylguanine-DNA methyltransferase (MGMT) is an independent predictor of therapeutic response and potential prognosis in patients with glioblastoma multiforme (GBM). However, its significance of clinical prognosis in different continents still needs to be explored. PATIENTS AND METHODS To explore the effects of MGMT promoter methylation on both progression-free survival (PFS) and overall survival (OS) among GBM patients from different continents, a systematic review of published studies was conducted. RESULTS A total of 5103 patients from 53 studies were involved in the systematic review and the total percentage of MGMT promoter methylation was 45.53%. Of these studies, 16 studies performed univariate analyses and 17 performed multivariate analyses of MGMT promoter methylation on PFS. The pooled hazard ratio (HR) estimated for PFS was 0.55 (95% CI 0.50, 0.60) by univariate analysis and 0.43 (95% CI 0.38, 0.48) by multivariate analysis. The effect of MGMT promoter methylation on OS was explored in 30 studies by univariate analysis and in 30 studies by multivariate analysis. The combined HR was 0.48 (95% CI 0.44, 0.52) and 0.42 (95% CI 0.38, 0.45), respectively. CONCLUSION In each subgroup divided by areas, the prognostic significance still remained highly significant. The proportion of methylation in each group was in inverse proportion to the corresponding HR in the univariate and multivariate analyses of PFS. However, from the perspective of OS, compared with data from Europe and the US, higher methylation rates in Asia did not bring better returns.
Collapse
Affiliation(s)
- Wei Meng
- Department of Pediatric Neurosurgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai
| | - Yangyang Jiang
- Department of Neurosurgery, Shanghai Deji Hospital, Shanghai Neuromedical Center Affiliated to Qingdao University, Shanghai, People's Republic of China
| | - Jie Ma
- Department of Pediatric Neurosurgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai
| |
Collapse
|
26
|
Wei KC, Chen CY, Feng LY, Huang WT, Chen CH, Hsu PW, Wang K, Hood LE, Chen LY. The rs16906252:C>T SNP is not associated with increased overall survival or temozolomide response in a Han-Chinese glioma cohort. PLoS One 2017; 12:e0178842. [PMID: 28575062 PMCID: PMC5456392 DOI: 10.1371/journal.pone.0178842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 05/19/2017] [Indexed: 12/31/2022] Open
Abstract
The methylation status of O-6-methylguanine-DNA methyltransferase (MGMT) is associated with the prognosis in gliomas and in other cancers. Recent studies showed that rs16906252, an SNP in the MGMT promoter, is associated with promoter methylation and is a predictor of the overall survival time (OST) and the response to temozolomide (TMZ) treatment. However, these findings haven’t been systematically investigated in the Han-Chinese population. We analyzed the relevance between rs16906252 polymorphisms, the MGMT methylation status, and the OST in 72 Han-Chinese gliomas patients. The MGMT promoter methylation was measured by bisulfite conversion followed by pyro-sequencing, while rs16906252 was measured by restriction endonuclease digestion. Contrary to the previous findings, we found no association between rs16906252 genotypes and promoter methylation on MGMT. The lower-grade glioma (LGGs) patients carrying the C allele with rs16906252 showed a surprisingly better OST (P = 0.04). Furthermore, the LGG patients carrying hypo-methylated MGMT promoter and rs16906252 T allele showed significantly poorer prognosis. The prognostic benefit of MGMT promoter methylation and genotypes on gliomas patients is marginal. A new molecular stratified patient grouping of LGGs is potentially associated with poorer OST. Active MGMT might have a protective role in LGG tumors, enabling evolution to severe malignancy.
Collapse
Affiliation(s)
- Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital-Linkou Medical Center, Taoyuan, Taiwan, Republic of China
- College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Chia-Yuan Chen
- Department of Medical Research and Development, Chang Gung Memorial Hospital-Linkou Medical Center, Taoyuan, Taiwan, Republic of China
| | - Li-Ying Feng
- Department of Neurosurgery, Chang Gung Memorial Hospital-Linkou Medical Center, Taoyuan, Taiwan, Republic of China
- College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Wei-Tzu Huang
- Department of Medical Research and Development, Chang Gung Memorial Hospital-Linkou Medical Center, Taoyuan, Taiwan, Republic of China
| | - Chia-Hua Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital-Linkou Medical Center, Taoyuan, Taiwan, Republic of China
| | - Peng-Wei Hsu
- Department of Neurosurgery, Chang Gung Memorial Hospital-Linkou Medical Center, Taoyuan, Taiwan, Republic of China
| | - Kai Wang
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Leroy E. Hood
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Leslie Y. Chen
- Department of Medical Research and Development, Chang Gung Memorial Hospital-Linkou Medical Center, Taoyuan, Taiwan, Republic of China
- Institute for Systems Biology, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
27
|
Wang HW, Xu ZK, Song Y, Liu YG. Correlations of MGMT genetic polymorphisms with temozolomide resistance and prognosis of patients with malignant gliomas: a population-based study in China. Cancer Gene Ther 2017; 24:215-220. [PMID: 28409559 DOI: 10.1038/cgt.2017.7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/16/2017] [Accepted: 02/18/2017] [Indexed: 12/22/2022]
Abstract
This study aims to investigate the associations of O6-methylguanine-DNA methyltransferase (MGMT) genetic polymorphisms (Leu84Phe and Ile143Val) with temozolomide (TMZ) resistance and prognosis of patients with malignant gliomas. A total of 212 patients diagnosed with malignant gliomas were enrolled in this study as the case group. All of these patients took oral TMZ and were assigned into the TMZ-sensitive (complete response+partial response) and the TMZ-resistant (stable disease+progressive disease) groups based on the clinical response after chemotherapy. The polymerase chain reaction-restriction fragment length polymorphism was used to identify the gene polymorphism of Leu84Phe and Ile143Val. The survival time and survival outcomes of all the patients were obtained by follow-up. There were significant differences in the genotype and allele of Leu84Phe between the TMZ-sensitive and the TMZ-resistant groups. The CT, TT and CT+TT genotypes and the T allele of MGMT gene Leu84Phe may be associated with increasing TMZ resistance in patients with malignant gliomas. Logistic regression analysis showed that Leu84Phe of MGMT gene and pathological grade were independent risk factors for the increase of TMZ resistance in patients with malignant gliomas. Kaplan-Meier survival curve revealed that the average survival time of patients with the CT+TT and CC genotypes of Leu84Phe in the two groups was statistically significant. COX regression analysis showed that Leu84Phe, degree of resection and pathological grade were independent prognostic factors for patients with malignant gliomas. Our study demonstrates that Leu84Phe of MGMT gene might be a risk factor of TMZ resistance and poor prognosis of patients with malignant gliomas.
Collapse
Affiliation(s)
- H-W Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Z-K Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Y Song
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Y-G Liu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| |
Collapse
|
28
|
Zhang J, Yang JH, Quan J, Kang X, Wang HJ, Dai PG. Identification of MGMT promoter methylation sites correlating with gene expression and IDH1 mutation in gliomas. Tumour Biol 2016; 37:13571-13579. [PMID: 27468718 DOI: 10.1007/s13277-016-5153-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 07/12/2016] [Indexed: 12/30/2022] Open
Abstract
O6-methylguanine-DNA methyltransferase (MGMT) gene promoter methylation was reported to be an independent prognostic and predictive factor in glioma patients who received temozolomide treatment. However, the predictive value of MGMT methylation was recently questioned by several large clinical studies. The purpose of this study is to identify MGMT gene promoter CpG sites or region whose methylation were closely correlated with its gene expression to elucidate this contradictory clinical observations. The methylation status for all CpG dinucleotides in MGMT promoter and first exon region were determined in 42 Chinese glioma patients, which were then correlated with MGMT gene expression, IDH1 mutation, and tumor grade. In whole 87 CpG dinucleotides analyzed, three distinct CpG regions covering 28 CpG dinucleotides were significantly correlated with MGMT gene expression; 10 CpG dinucleotides were significantly correlated with glioma classification (p < 0.05). Isocitrate dehydrogenase 1 (IDH1) mutation and MGMT gene hypermethylation significantly co-existed, but not for MGMT gene expression. The validation cohort of gliomas treated with standard of care and comparison of the CpGs we identified with the current CpGs used in clinical setting will be very important for gliomas individual medicine in the future.
Collapse
Affiliation(s)
- Jie Zhang
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, China
| | - Jian-Hui Yang
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, China
| | - Jia Quan
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, China
| | - Xing Kang
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, China
| | - Hui-Juan Wang
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, China
| | - Peng-Gao Dai
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, China.
| |
Collapse
|
29
|
MGMT DNA repair gene promoter/enhancer haplotypes alter transcription factor binding and gene expression. Cell Oncol (Dordr) 2016; 39:435-447. [PMID: 27306526 DOI: 10.1007/s13402-016-0286-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2016] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND The O6-methylguanine-DNA methyltransferase (MGMT) protein removes O6-alkyl-guanine adducts from DNA. MGMT expression can thus alter the sensitivity of cells and tissues to environmental and chemotherapeutic alkylating agents. Previously, we defined the haplotype structure encompassing single nucleotide polymorphisms (SNPs) in the MGMT promoter/enhancer (P/E) region and found that haplotypes, rather than individual SNPs, alter MGMT promoter activity. The exact mechanism(s) by which these haplotypes exert their effect on MGMT promoter activity is currently unknown, but we noted that many of the SNPs comprising the MGMT P/E haplotypes are located within or in close proximity to putative transcription factor binding sites. Thus, these haplotypes could potentially affect transcription factor binding and, subsequently, alter MGMT promoter activity. METHODS In this study, we test the hypothesis that MGMT P/E haplotypes affect MGMT promoter activity by altering transcription factor (TF) binding to the P/E region. We used a promoter binding TF profiling array and a reporter assay to evaluate the effect of different P/E haplotypes on TF binding and MGMT expression, respectively. RESULTS Our data revealed a significant difference in TF binding profiles between the different haplotypes evaluated. We identified TFs that consistently showed significant haplotype-dependent binding alterations (p ≤ 0.01) and revealed their role in regulating MGMT expression using siRNAs and a dual-luciferase reporter assay system. CONCLUSIONS The data generated support our hypothesis that promoter haplotypes alter the binding of TFs to the MGMT P/E and, subsequently, affect their regulatory function on MGMT promoter activity and expression level.
Collapse
|
30
|
Kuroiwa-Trzmielina J, Wang F, Rapkins RW, Ward RL, Buchanan DD, Win AK, Clendenning M, Rosty C, Southey MC, Winship IM, Hopper JL, Jenkins MA, Olivier J, Hawkins NJ, Hitchins MP. SNP rs16906252C>T Is an Expression and Methylation Quantitative Trait Locus Associated with an Increased Risk of Developing MGMT-Methylated Colorectal Cancer. Clin Cancer Res 2016; 22:6266-6277. [PMID: 27267851 DOI: 10.1158/1078-0432.ccr-15-2765] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 05/20/2016] [Accepted: 06/01/2016] [Indexed: 01/15/2023]
Abstract
PURPOSE Methylation of the MGMT promoter is the major cause of O6-methylguanine methyltransferase deficiency in cancer and has been associated with the T variant of the promoter enhancer SNP rs16906252C>T. We sought evidence for an association between the rs16906252C>T genotype and increased risk of developing a subtype of colorectal cancer featuring MGMT methylation, mediated by genotype-dependent epigenetic silencing within normal tissues. EXPERIMENTAL DESIGN By applying a molecular pathologic epidemiology case-control study design, associations between rs16906252C>T and risk for colorectal cancer overall, and colorectal cancer stratified by MGMT methylation status, were estimated using multinomial logistic regression in two independent retrospective series of colorectal cancer cases and controls. The test sample comprised 1,054 colorectal cancer cases and 451 controls from Sydney, Australia. The validation sample comprised 612 colorectal cancer cases and 245 controls from the Australasian Colon Cancer Family Registry (ACCFR). To determine whether rs16906252C>T was linked to a constitutively altered epigenetic state, quantitative allelic expression and methylation analyses were performed in normal tissues. RESULTS An association between rs16906252C>T and increased risk of developing MGMT-methylated colorectal cancer in the Sydney sample was observed [OR, 3.3; 95% confidence interval (CI), 2.0-5.3; P < 0.0001], which was replicated in the ACCFR sample (OR, 4.0; 95% CI, 2.4-6.8; P < 0.0001). The T allele demonstrated about 2.5-fold reduced transcription in normal colorectal mucosa from cases and controls and was selectively methylated in a minority of normal cells, indicating that rs16906252C>T represents an expression and methylation quantitative trait locus. CONCLUSIONS We provide evidence that rs16906252C>T is associated with elevated risk for MGMT-methylated colorectal cancer, likely mediated by constitutive epigenetic repression of the T allele. Clin Cancer Res; 22(24); 6266-77. ©2016 AACR.
Collapse
Affiliation(s)
- Joice Kuroiwa-Trzmielina
- Medical Epigenetics Laboratory, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Fan Wang
- Department of Medicine (Oncology), Stanford University, Stanford, California.,School of Public Health (Epidemiology), Harbin Medical University, Harbin, PR China
| | - Robert W Rapkins
- Medical Epigenetics Laboratory, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia.,Cure Brain Cancer Foundation Biomarkers and Translational Research Adult Cancer Program, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Robyn L Ward
- Integrated Cancer Research Group, Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Sydney, Australia
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Aung Ko Win
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Mark Clendenning
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Australia
| | - Christophe Rosty
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Australia.,Envoi Specialist Pathologists, Herston, Australia.,School of Medicine, University of Queensland, Herston, Australia
| | - Melissa C Southey
- Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Australia
| | - Ingrid M Winship
- Genetic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Australia.,Department of Medicine, The University of Melbourne, Parkville, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia.,Department of Epidemiology and Institute of Health and Environment, School of Public Health, Seoul National University, Seoul, Korea
| | - Mark A Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Jake Olivier
- School of Mathematics and Statistics, University of New South Wales, Sydney, Australia
| | - Nicholas J Hawkins
- Department of Pathology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Megan P Hitchins
- Department of Medicine (Oncology), Stanford University, Stanford, California.
| |
Collapse
|
31
|
Kristensen LS, Michaelsen SR, Dyrbye H, Aslan D, Grunnet K, Christensen IJ, Poulsen HS, Grønbæk K, Broholm H. Assessment of Quantitative and Allelic MGMT Methylation Patterns as a Prognostic Marker in Glioblastoma. J Neuropathol Exp Neurol 2016; 75:246-55. [PMID: 26883115 PMCID: PMC4777218 DOI: 10.1093/jnen/nlv024] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Methylation of the O6-methylguanine-DNA methyltransferase (MGMT) gene is a predictive and prognostic marker in newly diagnosed glioblastoma patients treated with temozolomide but how MGMT methylation should be assessed to ensure optimal detection accuracy is debated. We developed a novel quantitative methylation-specific PCR (qMSP) MGMT assay capable of providing allelic methylation data and analyzed 151 glioblastomas from patients receiving standard of care treatment (Stupp protocol). The samples were also analyzed by immunohistochemistry (IHC), standard bisulfite pyrosequencing, and genotyped for the rs1690252 MGMT promoter single nucleotide polymorphism. Monoallelic methylation was observed more frequently than biallelic methylation, and some cases with monoallelic methylation expressed the MGMT protein whereas others did not. The presence of MGMT methylation was associated with better overall survival (p = 0.006; qMSP and p = 0.002; standard pyrosequencing), and the presence of the protein was associated with worse overall survival (p = 0.009). Combined analyses of qMSP and standard pyrosequencing or IHC identified additional patients who benefited from temozolomide treatment. Finally, low methylation levels were also associated with better overall survival (p = 0.061; qMSP and p = 0.02; standard pyrosequencing). These data support the use of both MGMT methylation and MGMT IHC but not allelic methylation data as prognostic markers in patients with temozolomide-treated glioblastoma.
Collapse
Affiliation(s)
- Lasse S Kristensen
- From the Department of Hematology (LSK, DA, KG); Department of Radiation Biology (SRM, KG, IJC, HSP); and Department of Pathology, Rigshospitalet, Copenhagen Ø, Denmark (HD, HB).
| | - Signe R Michaelsen
- From the Department of Hematology (LSK, DA, KG); Department of Radiation Biology (SRM, KG, IJC, HSP); and Department of Pathology, Rigshospitalet, Copenhagen Ø, Denmark (HD, HB)
| | - Henrik Dyrbye
- From the Department of Hematology (LSK, DA, KG); Department of Radiation Biology (SRM, KG, IJC, HSP); and Department of Pathology, Rigshospitalet, Copenhagen Ø, Denmark (HD, HB)
| | - Derya Aslan
- From the Department of Hematology (LSK, DA, KG); Department of Radiation Biology (SRM, KG, IJC, HSP); and Department of Pathology, Rigshospitalet, Copenhagen Ø, Denmark (HD, HB)
| | - Kirsten Grunnet
- From the Department of Hematology (LSK, DA, KG); Department of Radiation Biology (SRM, KG, IJC, HSP); and Department of Pathology, Rigshospitalet, Copenhagen Ø, Denmark (HD, HB)
| | - Ib J Christensen
- From the Department of Hematology (LSK, DA, KG); Department of Radiation Biology (SRM, KG, IJC, HSP); and Department of Pathology, Rigshospitalet, Copenhagen Ø, Denmark (HD, HB)
| | - Hans S Poulsen
- From the Department of Hematology (LSK, DA, KG); Department of Radiation Biology (SRM, KG, IJC, HSP); and Department of Pathology, Rigshospitalet, Copenhagen Ø, Denmark (HD, HB)
| | - Kirsten Grønbæk
- From the Department of Hematology (LSK, DA, KG); Department of Radiation Biology (SRM, KG, IJC, HSP); and Department of Pathology, Rigshospitalet, Copenhagen Ø, Denmark (HD, HB)
| | - Helle Broholm
- From the Department of Hematology (LSK, DA, KG); Department of Radiation Biology (SRM, KG, IJC, HSP); and Department of Pathology, Rigshospitalet, Copenhagen Ø, Denmark (HD, HB)
| |
Collapse
|
32
|
Fogli A, Chautard E, Vaurs-Barrière C, Pereira B, Müller-Barthélémy M, Court F, Biau J, Pinto AA, Kémény JL, Khalil T, Karayan-Tapon L, Verrelle P, Costa BM, Arnaud P. The tumoral A genotype of the MGMT rs34180180 single-nucleotide polymorphism in aggressive gliomas is associated with shorter patients' survival. Carcinogenesis 2015; 37:169-176. [PMID: 26717998 DOI: 10.1093/carcin/bgv251] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/19/2015] [Indexed: 01/09/2023] Open
Abstract
Malignant gliomas are the most common primary brain tumors. Grade III and IV gliomas harboring wild-type IDH1/2 are the most aggressive. In addition to surgery and radiotherapy, concomitant and adjuvant chemotherapy with temozolomide (TMZ) significantly improves overall survival (OS). The methylation status of the O(6)-methylguanine-DNA methyltransferase (MGMT) promoter is predictive of TMZ response and a prognostic marker of cancer outcome. However, the promoter regions the methylation of which correlates best with survival in aggressive glioma and whether the promoter methylation status predictive value could be refined or improved by other MGMT-associated molecular markers are not precisely known. In a cohort of 87 malignant gliomas treated with radiotherapy and TMZ-based chemotherapy, we retrospectively determined the MGMT promoter methylation status, genotyped single nucleotide polymorphisms (SNPs) in the promoter region and quantified MGMT mRNA expression level. Each of these variables was correlated with each other and with the patients' OS. We found that methylation of the CpG sites within MGMT exon 1 best correlated with OS and MGMT expression levels, and confirmed MGMT methylation as a stronger independent prognostic factor compared to MGMT transcription levels. Our main finding is that the presence of only the A allele at the rs34180180 SNP in the tumor was significantly associated with shorter OS, independently of the MGMT methylation status. In conclusion, in the clinic, rs34180180 SNP genotyping could improve the prognostic value of the MGMT promoter methylation assay in patients with aggressive glioma treated with TMZ.
Collapse
Affiliation(s)
- Anne Fogli
- INSERM-U1103 and.,CNRS-UMR 6293, Clermont-Ferrand 63001, France.,GReD Laboratory, Clermont Auvergne University, Clermont-Ferrand 63000, France.,Biochemistry and Molecular Biology Department, Clermont-Ferrand Hospital, Clermont-Ferrand 63003, France
| | - Emmanuel Chautard
- Clermont Auvergne University, EA 7283 CREaT, Clermont-Ferrand 63000, France.,Radiotherapy Department, Jean Perrin Center, Clermont-Ferrand 63011, France
| | - Catherine Vaurs-Barrière
- INSERM-U1103 and.,CNRS-UMR 6293, Clermont-Ferrand 63001, France.,GReD Laboratory, Clermont Auvergne University, Clermont-Ferrand 63000, France
| | - Bruno Pereira
- Biostatistics Department , DRCI, Clermont-Ferrand Hospital , Clermont-Ferrand 63003 , France
| | | | - Franck Court
- INSERM-U1103 and.,CNRS-UMR 6293, Clermont-Ferrand 63001, France.,GReD Laboratory, Clermont Auvergne University, Clermont-Ferrand 63000, France
| | - Julian Biau
- Clermont Auvergne University, EA 7283 CREaT, Clermont-Ferrand 63000, France.,Radiotherapy Department, Jean Perrin Center, Clermont-Ferrand 63011, France
| | - Afonso Almeida Pinto
- Department of Neurosurgery , Braga Hospital , Braga 4710-243 São Victor , Portugal
| | - Jean-Louis Kémény
- Department of Anatomopathology , Clermont-Ferrand Hospital , Clermont-Ferrand 63003 , France
| | - Toufic Khalil
- Department of Neurosurgery, Clermont-Ferrand Hospital, Clermont-Ferrand 63003, France.,Clermont Auvergne University, EA 7282 IGCNC, Clermont-Ferrand 63000, France
| | - Lucie Karayan-Tapon
- INSERM-U935, Poitiers 86021, France.,Poitiers University, Poitiers 86000, France.,Cancer Biology Laboratory, Poitiers Hospital, Poitiers 86021, France
| | - Pierre Verrelle
- Clermont Auvergne University, EA 7283 CREaT, Clermont-Ferrand 63000, France.,Radiotherapy Department, Jean Perrin Center, Clermont-Ferrand 63011, France.,INSERM U2021 CNRS UMR3347, Curie Institute, Orsay 91405, France
| | - Bruno Marques Costa
- School of Health Sciences, Life and Health Sciences Research Institute (ICVS), Braga 4710-057, Portugal and.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Braga 4710-057, Portugal
| | - Philippe Arnaud
- INSERM-U1103 and.,CNRS-UMR 6293, Clermont-Ferrand 63001, France.,GReD Laboratory, Clermont Auvergne University, Clermont-Ferrand 63000, France
| |
Collapse
|
33
|
Lv JF, Hu L, Zhuo W, Zhang CM, Zhou HH, Fan L. Epigenetic alternations and cancer chemotherapy response. Cancer Chemother Pharmacol 2015; 77:673-84. [DOI: 10.1007/s00280-015-2951-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 12/17/2015] [Indexed: 12/29/2022]
|
34
|
Nishihara R, VanderWeele TJ, Shibuya K, Mittleman MA, Wang M, Field AE, Giovannucci E, Lochhead P, Ogino S. Molecular pathological epidemiology gives clues to paradoxical findings. Eur J Epidemiol 2015; 30:1129-35. [PMID: 26445996 PMCID: PMC4639412 DOI: 10.1007/s10654-015-0088-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/26/2015] [Indexed: 12/23/2022]
Abstract
A number of epidemiologic studies have described what appear to be paradoxical associations, where an incongruous relationship is observed between a certain well-established risk factor for disease incidence and favorable clinical outcome among patients with that disease. For example, the "obesity paradox" represents the association between obesity and better survival among patients with a certain disease such as coronary heart disease. Paradoxical observations cause vexing clinical and public health problems as they raise questions on causal relationships and hinder the development of effective interventions. Compelling evidence indicates that pathogenic processes encompass molecular alterations within cells and the microenvironment, influenced by various exogenous and endogenous exposures, and that interpersonal heterogeneity in molecular pathology and pathophysiology exists among patients with any given disease. In this article, we introduce methods of the emerging integrative interdisciplinary field of molecular pathological epidemiology (MPE), which is founded on the unique disease principle and disease continuum theory. We analyze and decipher apparent paradoxical findings, utilizing the MPE approach and available literature data on tumor somatic genetic and epigenetic characteristics. Through our analyses in colorectal cancer, renal cell carcinoma, and glioblastoma (malignant brain tumor), we can readily explain paradoxical associations between disease risk factors and better prognosis among disease patients. The MPE paradigm and approach can be applied to not only neoplasms but also various non-neoplastic diseases where there exists indisputable ubiquitous heterogeneity of pathogenesis and molecular pathology. The MPE paradigm including consideration of disease heterogeneity plays an essential role in advancements of precision medicine and public health.
Collapse
Affiliation(s)
- Reiko Nishihara
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Ave., Boston, MA, 02115, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA, 02215, USA.
- Department of Global Health Policy, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan.
| | - Tyler J VanderWeele
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA, 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA, 02115, USA
| | - Kenji Shibuya
- Department of Global Health Policy, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Murray A Mittleman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA, 02115, USA
- Cardiovascular Epidemiology Research Unit, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 375 Longwood Ave., Boston, MA, USA
| | - Molin Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA, 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA, 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Ave., Boston, MA, 02115, USA
| | - Alison E Field
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA, 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Ave., Boston, MA, 02115, USA
- Division of Adolescent Medicine, Boston Children's Hospital, 300 Longwood Ave., Boston, MA, 02115, USA
- Department of Epidemiology, Brown University, 121 South Main Street, Providence, RI, 02912, USA
| | - Edward Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 655 Huntington Ave., Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA, 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Ave., Boston, MA, 02115, USA
| | - Paul Lochhead
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Shuji Ogino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA, 02215, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA, 02115, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
35
|
Bienkowski M, Berghoff AS, Marosi C, Wöhrer A, Heinzl H, Hainfellner JA, Preusser M. Clinical Neuropathology practice guide 5-2015: MGMT methylation pyrosequencing in glioblastoma: unresolved issues and open questions. Clin Neuropathol 2015; 34:250-7. [PMID: 26295302 PMCID: PMC4542181 DOI: 10.5414/np300904] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 07/20/2015] [Indexed: 01/01/2023] Open
Abstract
O6-methylguanine-methyltransferase (MGMT) promoter methylation status has prognostic and, in the subpopulation of elderly patients, predictive value in newly diagnosed glioblastoma. Therefore, knowledge of the MGMT promoter methylation status is important for clinical decision-making. So far, MGMT testing has been limited by the lack of a robust test with sufficiently high analytical performance. Recently, one of several available pyrosequencing protocols has been shown to be an accurate and robust method for MGMT testing in an intra- and interlaboratory ring trial. However, some uncertainties remain with regard to methodological issues, cut-off definitions, and optimal use in the clinical setting. In this article, we highlight and discuss several of these open questions. The main unresolved issues are the definition of the most relevant CpG sites to analyze for clinical purposes and the determination of a cut-off value for dichotomization of quantitative MGMT pyrosequencing results into "MGMT methylated" and "MGMT unmethylated" patient subgroups as a basis for further treatment decisions.
Collapse
Affiliation(s)
- Michal Bienkowski
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
- Department of Molecular Pathology and Neuropathology, Medical University of Lodz, Lodz, Poland
| | - Anna S. Berghoff
- Department of Medicine I
- Comprehensive Cancer Center-CNS Tumours Unit (CCC-CNS), and
| | - Christine Marosi
- Department of Medicine I
- Comprehensive Cancer Center-CNS Tumours Unit (CCC-CNS), and
| | - Adelheid Wöhrer
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center-CNS Tumours Unit (CCC-CNS), and
| | - Harald Heinzl
- Comprehensive Cancer Center-CNS Tumours Unit (CCC-CNS), and
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Johannes A. Hainfellner
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center-CNS Tumours Unit (CCC-CNS), and
| | - Matthias Preusser
- Department of Medicine I
- Comprehensive Cancer Center-CNS Tumours Unit (CCC-CNS), and
| |
Collapse
|
36
|
Leng S, Wu G, Collins LB, Thomas CL, Tellez CS, Jauregui AR, Picchi MA, Zhang X, Juri DE, Desai D, Amin SG, Crowell RE, Stidley CA, Liu Y, Swenberg JA, Lin Y, Wathelet MG, Gilliland FD, Belinsky SA. Implication of a Chromosome 15q15.2 Locus in Regulating UBR1 and Predisposing Smokers to MGMT Methylation in Lung. Cancer Res 2015; 75:3108-17. [PMID: 26183928 DOI: 10.1158/0008-5472.can-15-0243] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/22/2015] [Indexed: 11/16/2022]
Abstract
O(6)-Methylguanine-DNA methyltransferase (MGMT) is a DNA repair enzyme that protects cells from carcinogenic effects of alkylating agents; however, MGMT is silenced by promoter hypermethylation during carcinogenesis. A single-nucleotide polymorphism (SNP) in an enhancer in the MGMT promoter was previously identified to be highly significantly associated with risk for MGMT methylation in lung cancer and sputum from smokers. To further genetic investigations, a genome-wide association and replication study was conducted in two smoker cohorts to identify novel loci for MGMT methylation in sputum that were independent of the MGMT enhancer polymorphism. Two novel trans-acting loci (15q15.2 and 17q24.3) that were identified acted together with the enhancer SNP to empower risk prediction for MGMT methylation. We found that the predisposition to MGMT methylation arising from the 15q15.2 locus involved regulation of the ubiquitin protein ligase E3 component UBR1. UBR1 attenuation reduced turnover of MGMT protein and increased repair of O6-methylguanine in nitrosomethylurea-treated human bronchial epithelial cells, while also reducing MGMT promoter activity and abolishing MGMT induction. Overall, our results substantiate reduced gene transcription as a major mechanism for predisposition to MGMT methylation in the lungs of smokers, and support the importance of UBR1 in regulating MGMT homeostasis and DNA repair of alkylated DNA adducts in cells.
Collapse
Affiliation(s)
- Shuguang Leng
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Guodong Wu
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Leonard B Collins
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Cynthia L Thomas
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Carmen S Tellez
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Andrew R Jauregui
- Lung Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Maria A Picchi
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Xiequn Zhang
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Daniel E Juri
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Dhimant Desai
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Shantu G Amin
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Richard E Crowell
- Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Christine A Stidley
- Department of Internal Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Yushi Liu
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - James A Swenberg
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yong Lin
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Marc G Wathelet
- Lung Fibrosis Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Frank D Gilliland
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Steven A Belinsky
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico.
| |
Collapse
|
37
|
Rapkins RW, Wang F, Nguyen HN, Cloughesy TF, Lai A, Ha W, Nowak AK, Hitchins MP, McDonald KL. The MGMT promoter SNP rs16906252 is a risk factor for MGMT methylation in glioblastoma and is predictive of response to temozolomide. Neuro Oncol 2015; 17:1589-98. [PMID: 25910840 DOI: 10.1093/neuonc/nov064] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 03/20/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Promoter methylation of O(6)-methylguanine-DNA methyltransferase (MGMT) is an important predictive biomarker in glioblastoma. The T variant of the MGMT promoter-enhancer single nucleotide polymorphism (SNP; rs16906252) has been associated with the presence of MGMT promoter methylation in other cancers. We examined the association of the T allele of rs16906252 with glioblastoma development, tumor MGMT methylation, MGMT protein expression, and survival outcomes. METHODS Two independent temozolomide-treated glioblastoma cohorts-one Australian (Australian Genomics and Clinical Outcomes of Glioma, n = 163) and the other American (University of California Los Angeles/Kaiser Permanente Los Angeles, n = 159)-were studied. Allelic bisulphite sequencing was used to determine if methylation was specific to the T allele. Additionally, we compared the incidence of the T allele between glioblastoma cases and matched controls to assess whether it was a risk factor for developing MGMT methylated glioblastoma. RESULTS Carriage of the T allele of the rs16906252 SNP was associated with both MGMT methylation and low MGMT protein expression and predicted significantly longer survival in temozolomide-treated patients with both MGMT methylated and nonmethylated glioblastoma. Methylation was linked to the T allele, inferring that the T variant plays a key role in the acquisition of MGMT methylation. Carriage of the T allele was associated with a significantly elevated risk of developing glioblastoma (adjusted odds ratio, 1.96; P = .013), increasing further when glioblastoma was classified by the presence of MGMT methylation (adjusted odds ratio, 2.86; P = .001). CONCLUSIONS The T allele of the rs16906252 SNP is a key determinant in the acquisition of MGMT methylation in glioblastoma. Temozolomide-treated patients with the rs16906252 T genotype have better survival, irrespective of tumor methylation status.
Collapse
Affiliation(s)
- Robert W Rapkins
- Cure Brain Cancer Neuro-oncology Laboratory, Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia (R.W.R., W.H., K.L.M.); Department of Medicine (Oncology), Stanford Cancer Institute, Stanford University, Stanford, California (F.W., M.P.H.); School of Public Health, Harbin Medical University, Harbin, People's Republic of China (F.W.); Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California (H.N.N., T.F.C., A.L.); School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (A.N.)
| | - Fan Wang
- Cure Brain Cancer Neuro-oncology Laboratory, Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia (R.W.R., W.H., K.L.M.); Department of Medicine (Oncology), Stanford Cancer Institute, Stanford University, Stanford, California (F.W., M.P.H.); School of Public Health, Harbin Medical University, Harbin, People's Republic of China (F.W.); Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California (H.N.N., T.F.C., A.L.); School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (A.N.)
| | - HuyTram N Nguyen
- Cure Brain Cancer Neuro-oncology Laboratory, Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia (R.W.R., W.H., K.L.M.); Department of Medicine (Oncology), Stanford Cancer Institute, Stanford University, Stanford, California (F.W., M.P.H.); School of Public Health, Harbin Medical University, Harbin, People's Republic of China (F.W.); Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California (H.N.N., T.F.C., A.L.); School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (A.N.)
| | - Timothy F Cloughesy
- Cure Brain Cancer Neuro-oncology Laboratory, Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia (R.W.R., W.H., K.L.M.); Department of Medicine (Oncology), Stanford Cancer Institute, Stanford University, Stanford, California (F.W., M.P.H.); School of Public Health, Harbin Medical University, Harbin, People's Republic of China (F.W.); Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California (H.N.N., T.F.C., A.L.); School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (A.N.)
| | - Albert Lai
- Cure Brain Cancer Neuro-oncology Laboratory, Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia (R.W.R., W.H., K.L.M.); Department of Medicine (Oncology), Stanford Cancer Institute, Stanford University, Stanford, California (F.W., M.P.H.); School of Public Health, Harbin Medical University, Harbin, People's Republic of China (F.W.); Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California (H.N.N., T.F.C., A.L.); School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (A.N.)
| | - Wendy Ha
- Cure Brain Cancer Neuro-oncology Laboratory, Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia (R.W.R., W.H., K.L.M.); Department of Medicine (Oncology), Stanford Cancer Institute, Stanford University, Stanford, California (F.W., M.P.H.); School of Public Health, Harbin Medical University, Harbin, People's Republic of China (F.W.); Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California (H.N.N., T.F.C., A.L.); School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (A.N.)
| | - Anna K Nowak
- Cure Brain Cancer Neuro-oncology Laboratory, Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia (R.W.R., W.H., K.L.M.); Department of Medicine (Oncology), Stanford Cancer Institute, Stanford University, Stanford, California (F.W., M.P.H.); School of Public Health, Harbin Medical University, Harbin, People's Republic of China (F.W.); Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California (H.N.N., T.F.C., A.L.); School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (A.N.)
| | - Megan P Hitchins
- Cure Brain Cancer Neuro-oncology Laboratory, Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia (R.W.R., W.H., K.L.M.); Department of Medicine (Oncology), Stanford Cancer Institute, Stanford University, Stanford, California (F.W., M.P.H.); School of Public Health, Harbin Medical University, Harbin, People's Republic of China (F.W.); Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California (H.N.N., T.F.C., A.L.); School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (A.N.)
| | - Kerrie L McDonald
- Cure Brain Cancer Neuro-oncology Laboratory, Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia (R.W.R., W.H., K.L.M.); Department of Medicine (Oncology), Stanford Cancer Institute, Stanford University, Stanford, California (F.W., M.P.H.); School of Public Health, Harbin Medical University, Harbin, People's Republic of China (F.W.); Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, California (H.N.N., T.F.C., A.L.); School of Medicine and Pharmacology, University of Western Australia, Perth, Australia (A.N.)
| |
Collapse
|
38
|
McDonald KL, Tabone T, Nowak AK, Erber WN. Somatic mutations in glioblastoma are associated with methylguanine-DNA methyltransferase methylation. Oncol Lett 2015; 9:2063-2067. [PMID: 26137013 DOI: 10.3892/ol.2015.2980] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 02/02/2015] [Indexed: 01/18/2023] Open
Abstract
The high level of methylguanine-DNA methyltransferase (MGMT) in glioblastoma is responsible for resistance to alkylating agents, such as temozolomide (TMZ). In glioblastomas with a methylated MGMT promoter, MGMT deficiency is presumed, resulting in an enhanced effect of TMZ. The aim of the present study was to investigate whether genomic alterations work synergistically with MGMT methylation status and contribute to the response to treatment and overall prognosis in glioblastoma. The current study included a cohort of 35 glioblastoma patients, with MGMT promoter methylation present in 48% of tumors. MGMT methylation was associated with significantly longer median survival (29.0 months) compared with patients without MGMT methylated tumors (12.0 months), as well as longer median time to progression following TMZ treatment (13.2 months, compared with 5.6 months for patients with an unmethylated MGMT status). In addition, somatic variants in hot spot exonic regions of 50 key cancer genes were examined in these glioblastomas. Non-synonymous mutations in methylated MGMT glioblastomas were four times higher compared with unmethylated MGMT glioblastomas. Furthermore, significantly increased frequencies of mutations in the TP53, CDKN2A, PTEN and PIK3CA genes were detected in MGMT methylated glioblastomas. The relative significance of these mutations, and their contribution to TMZ sensitivity, adjunct to MGMT methylation, require further investigation in a larger cohort.
Collapse
Affiliation(s)
- Kerrie L McDonald
- Cure Brain Cancer Neuro-oncology Group, Prince of Wales Clinical School, Lowy Cancer Research Centre C25, University of New South Wales, Sydney, NSW 2052, Australia
| | - Tania Tabone
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley WA 6009, Australia
| | - Anna K Nowak
- Harry Perkins Institute of Medical Research, Nedlands WA 6009, Australia
| | - Wendy N Erber
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley WA 6009, Australia
| |
Collapse
|
39
|
Yang H, Wei D, Yang K, Tang W, Luo Y, Zhang J. The prognosis of MGMT promoter methylation in glioblastoma patients of different race: a meta-analysis. Neurochem Res 2014; 39:2277-87. [PMID: 25230908 DOI: 10.1007/s11064-014-1435-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 09/05/2014] [Accepted: 09/08/2014] [Indexed: 11/30/2022]
Abstract
O(6)-Methylguanine-DNA methyltransferase (MGMT) is a DNA repair gene. Epigenetic silencing of the MGMT promoter methylation compromises DNA repair and has been associated with longer survival in patients with glioblastoma (GBM) who receive alkylating agents. But the prognostic of MGMT promoter methylation in GBM patients of different race is still ambiguous. Based on an univariate or multivariate analysis between different race (Caucasian and Asian), a meta-analysis of the effects of MGMT promoter methylation on both progression-free survival (PFS) and overall survival (OS) among GBM patients was conducted. A total of 6,309 patients from 50 studies were involved in the analysis. Random effect models were applied to estimate the pooled hazard ratio (HR) with 95 % confidence intervals (CIs) for GBM patients of different race prognosis, the Chi square-based Q test was used to test heterogeneity. Begg's (funnel plot method) and Egger's linear regression tests were adopted to check publication bias (a bias with regard to what is likely to be published, among what is available to be published). The HR value estimated for OS was 0.524 (95 % CI 0.428-0.640) by univariate analysis and 0.427 (95 % CI 0.355-0.513) by multivariate analysis in Caucasian. The HR value estimated for OS was 0.892 (95 % CI 0.469-1.698) by univariate analysis and 0.562 (95 % CI 0.394-0.804) by multivariate analysis in Asian. The HR value estimated for PFS was 0.526 (95 % CI 0.372-0.743) by univariate analysis and 0.437 (95 % CI 0.356-0.537) by multivariate analysis in Caucasian. The HR value estimated for PFS was 0.132 (95 % CI 0.006-3.027) by multivariate analysis in Asian. This data revealed that GBM patients with MGMT promoter methylation had longer OS and PFS by univariate or multivariate analysis in Caucasian regardless of therapeutic intervention. However, GBM patients with MGMT promoter methylation only had longer OS by multivariate analysis in Asian.
Collapse
Affiliation(s)
- Haiyu Yang
- Lab of Molecular Genetics of Aging and Tumor, Faculty of Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | | | | | | | | | | |
Collapse
|
40
|
Tang J, Xiong Y, Zhou HH, Chen XP. DNA methylation and personalized medicine. J Clin Pharm Ther 2014; 39:621-7. [PMID: 25230364 DOI: 10.1111/jcpt.12206] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 08/17/2014] [Indexed: 12/12/2022]
Affiliation(s)
- J. Tang
- Department of Clinical Pharmacology; Xiangya Hospital; Central South University; Changsha China
- Pharmacogenetics Research Institute; Institute of Clinical Pharmacology; Hunan Key laboratory of Pharmacogenetics; Central South University; Changsha China
| | - Y. Xiong
- Department of Clinical Pharmacology; Xiangya Hospital; Central South University; Changsha China
- Pharmacogenetics Research Institute; Institute of Clinical Pharmacology; Hunan Key laboratory of Pharmacogenetics; Central South University; Changsha China
| | - H.-H. Zhou
- Department of Clinical Pharmacology; Xiangya Hospital; Central South University; Changsha China
- Pharmacogenetics Research Institute; Institute of Clinical Pharmacology; Hunan Key laboratory of Pharmacogenetics; Central South University; Changsha China
| | - X.-P. Chen
- Department of Clinical Pharmacology; Xiangya Hospital; Central South University; Changsha China
- Pharmacogenetics Research Institute; Institute of Clinical Pharmacology; Hunan Key laboratory of Pharmacogenetics; Central South University; Changsha China
| |
Collapse
|
41
|
Kanemoto M, Shirahata M, Nakauma A, Nakanishi K, Taniguchi K, Kukita Y, Arakawa Y, Miyamoto S, Kato K. Prognostic prediction of glioblastoma by quantitative assessment of the methylation status of the entire MGMT promoter region. BMC Cancer 2014; 14:641. [PMID: 25175833 PMCID: PMC4161852 DOI: 10.1186/1471-2407-14-641] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 08/27/2014] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation is reported to be a prognostic and predictive factor of alkylating chemotherapy for glioblastoma patients. Methylation specific PCR (MSP) has been most commonly used when the methylation status of MGMT is assessed. However, technical obstacles have hampered the implementation of MSP-based diagnostic tests. We quantitatively analyzed the methylation status of the entire MGMT promoter region and applied this information for prognostic prediction using sequencing technology. METHODS Between 1998 and 2012, the genomic DNA of 85 tumor samples from newly diagnosed glioblastoma patients was subjected to bisulfite treatment and subdivided into a training set, consisting of fifty-three samples, and a test set, consisting of thirty-two samples. The training set was analyzed by deep Sanger sequencing with a sequencing coverage of up to 96 clones per sample. This analysis quantitatively revealed the degree of methylation of each cytidine phosphate guanosine (CpG) site. Based on these data, we constructed a prognostic prediction system for glioblastoma patients using a supervised learning method. We then validated this prediction system by deep sequencing with a next-generation sequencer using a test set of 32 samples. RESULTS The methylation status of the MGMT promoter was correlated with progression-free survival (PFS) in our patient population in the training set. The degree of correlation differed among the CpG sites. Using the data from the top twenty CpG sites, we constructed a prediction system for overall survival (OS) and PFS. The system successfully classified patients into good and poor prognosis groups in both the training set (OS, p = 0.0381; PFS, p = 0.00122) and the test set (OS, p = 0.0476; PFS, p = 0.0376). Conventional MSP could not predict the prognosis in either of our sets. (training set: OS; p = 0.993 PFS; p = 0.113, test set: OS; p = 0.326 PFS; p = 0.342). CONCLUSIONS The prognostic ability of our prediction system using sequencing data was better than that of methylation-specific PCR (MSP). Advances in sequencing technologies will make this approach a plausible option for diagnoses based on MGMT promotor methylation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kikuya Kato
- Research Institute, Osaka Medical Center for Cancer and Cardiovascular Diseases, 1-3-3 Nakamichi, Higashinari-ku, Osaka, Japan.
| |
Collapse
|
42
|
Association of single-nucleotide polymorphisms in ERCC1 and ERCC2 with glioma risk. Tumour Biol 2014; 35:7451-7. [PMID: 24782032 DOI: 10.1007/s13277-014-1969-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/11/2014] [Indexed: 12/31/2022] Open
Abstract
We conducted a case-control study to assess the role of three single-nucleotide polymorphisms (SNPs) in excision repair cross-complementation group 1 (ERCC1) and two SNPs in excision repair cross-complementation group 2 (ERCC2) on the glioma risk in a Chinese population, and investigate the gene-environmental interaction for the cancer risk. A 1:2 matched case-control study was conducted. Logistic regression analysis revealed that individuals carrying ERCC1 rs2298881 CC genotype were associated with risk of glioma when compared with AA genotype carriers. The significant associations of ERCC1 rs2298881 polymorphism with glioma susceptibility were observed in both the dominant and the recessive models. In a stratification analysis, we found that ERCC1 rs2298881 variants showed an increased association with the risk of glioma in males, ever smokers, and high-grade glioma cases. In conclusion, our study suggests that ERCC1 rs2298881 polymorphism is associated with risk of glioma in codominant, dominant, and recessive models, especially in males, smokers, and high-grade glioma cases. This finding could be useful in revealing the genetic characteristics of glioma and suggests more effective strategies for prevention and treatment.
Collapse
|
43
|
Tabone T, Abuhusain HJ, Nowak AK, Erber WN, McDonald KL. Multigene profiling to identify alternative treatment options for glioblastoma: a pilot study. J Clin Pathol 2014; 67:550-5. [PMID: 24695838 DOI: 10.1136/jclinpath-2014-202173] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
UNLABELLED Glioblastoma (GBM) is a highly aggressive malignancy and the most effective treatment regime has a high relapse rate. Increasingly, the development of therapies involves defining drug-diagnostic combinations where the presence of a molecular target or marker identifies patients who are most likely to respond to a specific therapy. Trials in other solid cancers have demonstrated clear utility in the incorporation of biomarkers to stratify patients to targeted treatment, however, there are no mutations that are currently used to inform treatment options for GBM. AIMS We piloted the use of high-throughput next-generation sequencing technology to identify genetic mutations in 44 GBM specimens that may be amenable to current or future targeted therapeutic strategies. METHOD Somatic mutation profiling was performed using the AmpliSeq Cancer Hotspot Panel v2 and semiconductor sequencing technology. RESULTS A total of 66 mutations were detected in 35/44 (80%) patients. The number of mutations per tumour ranged from 0 to 4 (average per tumour=1.5). The most frequent mutations were in TP53 (n=12), PTEN (n=9), EGFR (n=8) and PIK3CA (n=5). Clinically actionable somatic mutations were detected in 24/35 (69%) patients. CONCLUSIONS This study demonstrates that the use of an 'off-the-shelf' oncogene primer panel and benchtop next-generation sequencer can identify mutations and potentially actionable targets in the majority of GBM patients. Data from this pilot highlights the potential for targeted genetic resequencing to identify mutations that may inform treatment options and predict outcomes.
Collapse
Affiliation(s)
- Tania Tabone
- Translational Cancer Pathology Laboratory, School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia, Australia
| | - Hazem J Abuhusain
- Cure Brain Cancer Neuro-oncology Group, Prince of Wales Clinical School, UNSW Australia
| | - Anna K Nowak
- Department of Medical Oncology, School of Medicine and Pharmacology, University of Western Australia, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | | | - Wendy N Erber
- Translational Cancer Pathology Laboratory, School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia, Australia
| | - Kerrie L McDonald
- Cure Brain Cancer Neuro-oncology Group, Prince of Wales Clinical School, UNSW Australia
| |
Collapse
|
44
|
Xu M, Nekhayeva I, Cross CE, Rondelli CM, Wickliffe JK, Abdel-Rahman SZ. Influence of promoter/enhancer region haplotypes on MGMT transcriptional regulation: a potential biomarker for human sensitivity to alkylating agents. Carcinogenesis 2014; 35:564-71. [PMID: 24163400 PMCID: PMC3941746 DOI: 10.1093/carcin/bgt355] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/04/2013] [Accepted: 10/18/2013] [Indexed: 01/01/2023] Open
Abstract
The O6-methylguanine-DNA methyltransferase gene (MGMT) encodes the direct reversal DNA repair protein that removes alkyl adducts from the O6 position of guanine. Several single-nucleotide polymorphisms (SNPs) exist in the MGMT promoter/enhancer (P/E) region. However, the haplotype structure encompassing these SNPs and their functional/biological significance are currently unknown. We hypothesized that MGMT P/E haplotypes, rather than individual SNPs, alter MGMT transcription and can thus alter human sensitivity to alkylating agents. To identify the haplotype structure encompassing the MGMT P/E region SNPs, we sequenced 104 DNA samples from healthy individuals and inferred the haplotypes using the data generated. We identified eight SNPs in this region, namely T7C (rs180989103), T135G (rs1711646), G290A (rs61859810), C485A (rs1625649), C575A (rs113813075), G666A (rs34180180), C777A (rs34138162) and C1099T (rs16906252). Phylogenetics and Sequence Evolution analysis predicted 21 potential haplotypes that encompass these SNPs ranging in frequencies from 0.000048 to 0.39. Of these, 10 were identified in our study population as 20 paired haplotype combinations. To determine the functional significance of these haplotypes, luciferase reporter constructs representing these haplotypes were transfected into glioblastoma cells and their effect on MGMT promoter activity was determined. Compared with the most common (reference) haplotype 1, seven haplotypes significantly upregulated MGMT promoter activity (18-119% increase; P < 0.05), six significantly downregulated MGMT promoter activity (29-97% decrease; P < 0.05) and one haplotype had no effect. Mechanistic studies conducted support the conclusion that MGMT P/E haplotypes, rather than individual SNPs, differentially regulate MGMT transcription and could thus play a significant role in human sensitivity to environmental and therapeutic alkylating agents.
Collapse
Affiliation(s)
- Meixiang Xu
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX 77555-1066, USA and
- Department of Global Environmental Health Sciences, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Ilona Nekhayeva
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX 77555-1066, USA and
- Department of Global Environmental Health Sciences, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Courtney E. Cross
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX 77555-1066, USA and
- Department of Global Environmental Health Sciences, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Catherine M. Rondelli
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX 77555-1066, USA and
- Department of Global Environmental Health Sciences, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | - Jeffrey K. Wickliffe
- Department of Global Environmental Health Sciences, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| | | |
Collapse
|
45
|
Oberstadt MC, Bien-Möller S, Weitmann K, Herzog S, Hentschel K, Rimmbach C, Vogelgesang S, Balz E, Fink M, Michael H, Zeden JP, Bruckmüller H, Werk AN, Cascorbi I, Hoffmann W, Rosskopf D, Schroeder HWS, Kroemer HK. Epigenetic modulation of the drug resistance genes MGMT, ABCB1 and ABCG2 in glioblastoma multiforme. BMC Cancer 2013; 13:617. [PMID: 24380367 PMCID: PMC3890604 DOI: 10.1186/1471-2407-13-617] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 12/20/2013] [Indexed: 12/19/2022] Open
Abstract
Background Resistance of the highly aggressive glioblastoma multiforme (GBM) to drug therapy is a major clinical problem resulting in a poor patient’s prognosis. Beside promoter methylation of the O6-methylguanine-DNA-methyltransferase (MGMT) gene the efflux transporters ABCB1 and ABCG2 have been suggested as pivotal factors contributing to drug resistance, but the methylation of ABCB1 and ABCG2 has not been assessed before in GBM. Methods Therefore, we evaluated the proportion and prognostic significance of promoter methylation of MGMT, ABCB1 and ABCG2 in 64 GBM patient samples using pyrosequencing technology. Further, the single nucleotide polymorphisms MGMT C-56 T (rs16906252), ABCB1 C3435T (rs1045642) and ABCG2 C421A (rs2231142) were determined using the restriction fragment length polymorphism method (RFLP). To study a correlation between promoter methylation and gene expression, we analyzed MGMT, ABCB1 and ABCG2 expression in 20 glioblastoma and 7 non-neoplastic brain samples. Results Despite a significantly increased MGMT and ABCB1 promoter methylation in GBM tissue, multivariate regression analysis revealed no significant association between overall survival of glioblastoma patients and MGMT or ABCB1 promoter methylation. However, a significant negative correlation between promoter methylation and expression could be identified for MGMT but not for ABCB1 and ABCG2. Furthermore, MGMT promoter methylation was significantly associated with the genotypes of the MGMT C-56 T polymorphism showing a higher methylation level in the T allele bearing GBM. Conclusions In summary, the data of this study confirm the previous published relation of MGMT promoter methylation and gene expression, but argue for no pivotal role of MGMT, ABCB1 and ABCG2 promoter methylation in GBM patients’ survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Heyo K Kroemer
- Department of Pharmacology, Ernst-Moritz-Arndt-University, Greifswald, Germany.
| |
Collapse
|
46
|
Wang F, Zhang S, Wen Y, Wei Y, Yan H, Liu H, Su J, Zhang Y, Che J. Revealing the architecture of genetic and epigenetic regulation: a maximum likelihood model. Brief Bioinform 2013; 15:1028-43. [DOI: 10.1093/bib/bbt076] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
47
|
Kristensen LS, Treppendahl MB, Asmar F, Girkov MS, Nielsen HM, Kjeldsen TE, Ralfkiaer E, Hansen LL, Grønbæk K. Investigation of MGMT and DAPK1 methylation patterns in diffuse large B-cell lymphoma using allelic MSP-pyrosequencing. Sci Rep 2013; 3:2789. [PMID: 24071855 PMCID: PMC3784959 DOI: 10.1038/srep02789] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 09/11/2013] [Indexed: 12/11/2022] Open
Abstract
The tumor suppressor genes MGMT and DAPK1 become methylated in several cancers including diffuse large B-cell lymphoma (DLBCL). However, allelic methylation patterns have not been investigated in DLBCL. We developed a fast and cost-efficient method for the analysis of allelic methylation based on pyrosequencing of methylation specific PCR (MSP) products including a SNP. Allelic methylation patterns were reliably analyzed in standards of known allelic methylation status even when diluted in unmethylated DNA to below 1% methylation. When studying 148 DLBCL patients MGMT and DAPK1 methylation was observed in 19% and 89%, respectively, and among methylated and heterozygous patients 29% and 55%, respectively, were biallelically methylated. An association between the T-allele of the rs16906252 SNP and MGMT methylation was observed (p-value=0.04), and DAPK1 methylation of the A-allele was associated with shorter overall survival (p-value=0.006). In future cancer research allelic MSP-pyrosequencing may be used to study a wide range of other loci.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Alleles
- Antibodies, Monoclonal, Murine-Derived
- Antineoplastic Combined Chemotherapy Protocols
- Cell Line
- Cyclophosphamide
- DNA Methylation
- DNA Modification Methylases/genetics
- DNA Repair Enzymes/genetics
- Death-Associated Protein Kinases/genetics
- Doxorubicin
- Female
- Genotype
- Humans
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Middle Aged
- Neoplasm Staging
- Polymorphism, Single Nucleotide
- Prednisone
- Rituximab
- Sensitivity and Specificity
- Sequence Analysis, DNA
- Treatment Outcome
- Tumor Suppressor Proteins/genetics
- Vincristine
- Young Adult
Collapse
Affiliation(s)
| | | | - Fazila Asmar
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | | | - Helene Myrtue Nielsen
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | | | - Kirsten Grønbæk
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
48
|
Jiang D, Zheng D, Wang L, Huang Y, Liu H, Xu L, Liao Q, Liu P, Shi X, Wang Z, Sun L, Zhou Q, Li N, Xu L, Le Y, Ye M, Shao G, Duan S. Elevated PLA2G7 gene promoter methylation as a gender-specific marker of aging increases the risk of coronary heart disease in females. PLoS One 2013; 8:e59752. [PMID: 23555769 PMCID: PMC3610900 DOI: 10.1371/journal.pone.0059752] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 02/18/2013] [Indexed: 12/22/2022] Open
Abstract
PLA2G7 gene product is a secreted enzyme whose activity is associated with coronary heart disease (CHD). The goal of our study is to investigate the contribution of PLA2G7 promoter DNA methylation to the risk of CHD. Using the bisulphite pyrosequencing technology, PLA2G7 methylation was measured among 36 CHD cases and 36 well-matched controls. Our results indicated that there was a significant association between PLA2G7 methylation and CHD (adjusted P = 0.025). Significant gender-specific correlation was observed between age and PLA2G7 methylation (males: adjusted r = −0.365, adjusted P = 0.037; females: adjusted r = 0.373, adjusted P = 0.035). A breakdown analysis by gender showed that PLA2G7 methylation was significantly associated with CHD in females (adjusted P = 0.003) but not in males. A further two-way ANOVA analysis showed there was a significant interaction between gender and status of CHD for PLA2G7 methylation (gender*CHD: P = 6.04E−7). Moreover, PLA2G7 methylation is associated with the levels of total cholesterols (TC, r = 0.462, P = 0.009), triglyceride (TG, r = 0.414, P = 0.02) and Apolipoprotein B (ApoB, r = 0.396, P = 0.028) in females but not in males (adjusted P>0.4). Receiver operating characteristic (ROC) curves showed that PLA2G7 methylation could predict the risk of CHD in females (area under curve (AUC) = 0.912, P = 2.40E−5). Our results suggest that PLA2G7 methylation changes with aging in a gender-specific pattern. The correlation between PLA2G7 methylation and CHD risk in females is independent of other parameters including age, smoking, diabetes and hypertension. PLA2G7 methylation might exert its effects on the risk of CHD by regulating the levels of TC, TG, and ApoB in females. The gender disparities in the PLA2G7 methylation may play a role in the molecular mechanisms underlying the pathophysiology of CHD.
Collapse
Affiliation(s)
- Danjie Jiang
- Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- The Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
| | - Dawei Zheng
- Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Lingyan Wang
- Bank of Blood Products, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Yi Huang
- The Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
| | - Haibo Liu
- Yinzhou People’s Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Leiting Xu
- The Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
| | - Qi Liao
- The Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
| | - Panpan Liu
- The Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
| | - Xinbao Shi
- Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Zhaoyang Wang
- Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Lebo Sun
- Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Qingyun Zhou
- Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Ni Li
- Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Limin Xu
- The Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
| | - Yanping Le
- The Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
| | - Meng Ye
- The Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
- * E-mail: (SD); (GS); (MY)
| | - Guofeng Shao
- Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- * E-mail: (SD); (GS); (MY)
| | - Shiwei Duan
- The Affiliated Hospital, Ningbo University, Ningbo, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
- * E-mail: (SD); (GS); (MY)
| |
Collapse
|