1
|
Bomsztyk K, Mar D, Denisenko O, Powell S, Vishnoi M, Yin Z, Delegard J, Hadley C, Tandon N, Patel AJ, Patel AP, Ellenbogen RG, Ramakrishna R, Rostomily RC. Analysis of DNA Methylation in Gliomas: Assessment of Preanalytical Variables. J Transl Med 2024; 104:102160. [PMID: 39426568 PMCID: PMC11709230 DOI: 10.1016/j.labinv.2024.102160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
Precision oncology is driven by biomarkers. For glioblastoma multiforme (GBM), the most common malignant adult primary brain tumor, O6-methylguanine-DNA methyltransferase (MGMT) gene promoter methylation is an important prognostic and treatment clinical biomarker. Time-consuming preanalytical steps such as biospecimen storage, fixation, sampling, and processing are sources of data irreproducibility, and all these preanalytical variables are confounded by intratumor heterogeneity of MGMT promoter methylation. To assess the effect of preanalytical variables on GBM DNA methylation, tissue storage/sampling (CryoGrid), sample preparation multisonicator (PIXUL), and 5-methylcytosine DNA immunoprecipitation (Matrix-MeDIP-qPCR/seq) platforms were used. MGMT promoter methylation status assayed by MeDIP-qPCR was validated with methylation-specific polymerase chain reaction. MGMT promoter methylation levels in frozen and formalin-fixed paraffin-embedded sample pairs were not statistically different, confirming the reliability of formalin-fixed paraffin-embedded for MGMT promoter methylation analysis. Warm ex vivo ischemia (up to 4 hours at 37 °C) and 3 cycles of repeated sample thawing and freezing did not statistically impact 5-methylcytosine at MGMT promoter, exon, and enhancer regions, indicating the resistance of DNA methylation to common variations in sample processing conditions that might be encountered in research and clinical settings. Twenty-six percent to 34% of specimens exhibited intratumor heterogeneity in the MGMT DNA promoter methylation. These data demonstrate that variations in sample fixation, ischemia duration and temperature, and DNA methylation assay technique do not have a statistically significant impact on MGMT promoter methylation assessment. However, intratumor methylation heterogeneity underscores the value of multiple biopsies at different GBM geographic tumor sites in the evaluation of MGMT promoter methylation status. Matrix-MeDIP-seq analysis revealed that MGMT promoter methylation status clustered with other differentially methylated genomic loci (eg, HOXA and lncRNAs) that are resilient to variation in the above preanalytical conditions. These observations offer new opportunities to develop more granular data-based epigenetic GBM biomarkers. In this regard, the high-throughput CryoGrid-PIXUL-Matrix toolbox could be useful.
Collapse
Affiliation(s)
- Karol Bomsztyk
- UW Medicine South Lake Union, University of Washington, Seattle, Washington; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington; Matchstick Technologies, Inc, Kirkland, Washington.
| | - Daniel Mar
- UW Medicine South Lake Union, University of Washington, Seattle, Washington; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington
| | - Oleg Denisenko
- UW Medicine South Lake Union, University of Washington, Seattle, Washington
| | - Suzanne Powell
- Department of Neuropathology, Houston Methodist Hospital, Houston, Texas
| | - Monika Vishnoi
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, Texas
| | - Zheng Yin
- Department of Systems Medicine and Bioengineering, Houston Methodist Neil Cancer Center, Houston, Texas
| | - Jennifer Delegard
- Department of Neurological Surgery, University of Washington, Seattle, Washington
| | - Caroline Hadley
- Department of Neurosurgery, University of New Mexico, Albuquerque, New Mexico
| | - Nitin Tandon
- Department of Neurosurgery, McGovern Medical School at UT Health, Houston, Texas
| | - Akash J Patel
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Anoop P Patel
- Department of Neurosurgery, Duke University, Durham, North Carolina
| | - Richard G Ellenbogen
- Department of Neurological Surgery, University of Washington, Seattle, Washington
| | - Rohan Ramakrishna
- Department of Neurological Surgery, Weill Cornell Medicine, New York, New York
| | - Robert C Rostomily
- Department of Neurosurgery, Houston Methodist Research Institute, Houston, Texas; Department of Neurological Surgery, University of Washington, Seattle, Washington; Department of Neurological Surgery, Weill Cornell Medicine, New York, New York
| |
Collapse
|
2
|
Singh G, Singh A, Bae J, Manjila S, Spektor V, Prasanna P, Lignelli A. -New frontiers in domain-inspired radiomics and radiogenomics: increasing role of molecular diagnostics in CNS tumor classification and grading following WHO CNS-5 updates. Cancer Imaging 2024; 24:133. [PMID: 39375809 PMCID: PMC11460168 DOI: 10.1186/s40644-024-00769-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 08/31/2024] [Indexed: 10/09/2024] Open
Abstract
Gliomas and Glioblastomas represent a significant portion of central nervous system (CNS) tumors associated with high mortality rates and variable prognosis. In 2021, the World Health Organization (WHO) updated its Glioma classification criteria, most notably incorporating molecular markers including CDKN2A/B homozygous deletion, TERT promoter mutation, EGFR amplification, + 7/-10 chromosome copy number changes, and others into the grading and classification of adult and pediatric Gliomas. The inclusion of these markers and the corresponding introduction of new Glioma subtypes has allowed for more specific tailoring of clinical interventions and has inspired a new wave of Radiogenomic studies seeking to leverage medical imaging information to explore the diagnostic and prognostic implications of these new biomarkers. Radiomics, deep learning, and combined approaches have enabled the development of powerful computational tools for MRI analysis correlating imaging characteristics with various molecular biomarkers integrated into the updated WHO CNS-5 guidelines. Recent studies have leveraged these methods to accurately classify Gliomas in accordance with these updated molecular-based criteria based solely on non-invasive MRI, demonstrating the great promise of Radiogenomic tools. In this review, we explore the relative benefits and drawbacks of these computational frameworks and highlight the technical and clinical innovations presented by recent studies in the landscape of fast evolving molecular-based Glioma subtyping. Furthermore, the potential benefits and challenges of incorporating these tools into routine radiological workflows, aiming to enhance patient care and optimize clinical outcomes in the evolving field of CNS tumor management, have been highlighted.
Collapse
Affiliation(s)
- Gagandeep Singh
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA.
| | - Annie Singh
- Atal Bihari Vajpayee Institute of Medical Sciences, New Delhi, India
| | - Joseph Bae
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, USA
| | - Sunil Manjila
- Department of Neurological Surgery, Garden City Hospital, Garden City, MI, USA
| | - Vadim Spektor
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA
| | - Prateek Prasanna
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, USA
| | - Angela Lignelli
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
3
|
Galbraith K, Snuderl M. Molecular Pathology of Gliomas. Clin Lab Med 2024; 44:149-159. [PMID: 38821638 DOI: 10.1016/j.cll.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Gliomas are the most common adult and pediatric primary brain tumors. Molecular studies have identified features that can enhance diagnosis and provide biomarkers. IDH1/2 mutation with ATRX and TP53 mutations defines diffuse astrocytomas, whereas IDH1/2 mutations with 1p19q loss defines oligodendroglioma. Focal amplifications of receptor tyrosine kinase genes, TERT promoter mutation, and loss of chromosomes 10 and 13 with trisomy of chromosome 7 are characteristic features of glioblastoma and can be used for diagnosis. BRAF gene fusions and mutations in low-grade gliomas and histone H3 mutations in high-grade gliomas also can be used for diagnostics.
Collapse
Affiliation(s)
- Kristyn Galbraith
- Department of Pathology, NYU Langone Medical Center, 240 East 38th Street, 22nd Floor, New York, NY 10016, USA
| | - Matija Snuderl
- Department of Pathology, NYU Langone Medical Center, 240 East 38th Street, 22nd Floor, New York, NY 10016, USA.
| |
Collapse
|
4
|
Bomsztyk K, Mar D, Denisenko O, Powell S, Vishnoi M, Delegard J, Patel A, Ellenbogen RG, Ramakrishna R, Rostomily R. Analysis of gliomas DNA methylation: Assessment of pre-analytical variables. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586350. [PMID: 38586048 PMCID: PMC10996653 DOI: 10.1101/2024.03.26.586350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Precision oncology is driven by molecular biomarkers. For glioblastoma multiforme (GBM), the most common malignant adult primary brain tumor, O6-methylguanine-DNA methyltransferase ( MGMT ) gene DNA promoter methylation is an important prognostic and treatment clinical biomarker. Time consuming pre-analytical steps such as biospecimen storage before fixing, sampling, and processing are major sources of errors and batch effects, that are further confounded by intra-tumor heterogeneity of MGMT promoter methylation. To assess the effect of pre-analytical variables on GBM DNA methylation, tissue storage/sampling (CryoGrid), sample preparation multi-sonicator (PIXUL) and 5-methylcytosine (5mC) DNA immunoprecipitation (Matrix MeDIP-qPCR/seq) platforms were used. MGMT promoter CpG methylation was examined in 173 surgical samples from 90 individuals, 50 of these were used for intra-tumor heterogeneity studies. MGMT promoter methylation levels in paired frozen and formalin fixed paraffin embedded (FFPE) samples were very close, confirming suitability of FFPE for MGMT promoter methylation analysis in clinical settings. Matrix MeDIP-qPCR yielded similar results to methylation specific PCR (MS-PCR). Warm ex-vivo ischemia (37°C up to 4hrs) and 3 cycles of repeated sample thawing and freezing did not alter 5mC levels at MGMT promoter, exon and upstream enhancer regions, demonstrating the resistance of DNA methylation to the most common variations in sample processing conditions that might be encountered in research and clinical settings. 20-30% of specimens exhibited intratumor heterogeneity in the MGMT DNA promoter methylation. Collectively these data demonstrate that variations in sample fixation, ischemia duration and temperature, and DNA methylation assay technique do not have significant impact on assessment of MGMT promoter methylation status. However, intratumor methylation heterogeneity underscores the need for histologic verification and value of multiple biopsies at different GBM geographic tumor sites in assessment of MGMT promoter methylation. Matrix-MeDIP-seq analysis revealed that MGMT promoter methylation status clustered with other differentially methylated genomic loci (e.g. HOXA and lncRNAs), that are likewise resilient to variation in above post-resection pre-analytical conditions. These MGMT -associated global DNA methylation patterns offer new opportunities to validate more granular data-based epigenetic GBM clinical biomarkers where the CryoGrid-PIXUL-Matrix toolbox could prove to be useful.
Collapse
|
5
|
Baddam SR, Kalagara S, Kuna K, Enaganti S. Recent advancements and theranostics strategies in glioblastoma therapy. Biomed Mater 2023; 18:052007. [PMID: 37582381 DOI: 10.1088/1748-605x/acf0ab] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/15/2023] [Indexed: 08/17/2023]
Abstract
Glioblastoma (GBM) is the most aggressive and lethal malignant brain tumor, and it is challenging to cure with surgery and treatment. The prevention of permanent brain damage and tumor invasion, which is the ultimate cause of recurrence, are major obstacles in GBM treatment. Besides, emerging treatment modalities and newer genetic findings are helping to understand and manage GBM in patients. Accordingly, researchers are focusing on advanced nanomaterials-based strategies for tackling the various problems associated with GBM. In this context, researchers explored novel strategies with various alternative treatment approaches such as early detection techniques and theranostics approaches. In this review, we have emphasized the recent advancement of GBM cellular models and their roles in designing GBM therapeutics. We have added a special emphasis on the novel genetic and drug target findings as well as strategies for early detection. Besides, we have discussed various theranostic approaches such as hyperthermia therapy, phototherapy and image-guided therapy. Approaches utilized for targeted drug delivery to the GBM were also discussed. This article also describes the recentin vivo, in vitroandex vivoadvances using innovative theranostic approaches.
Collapse
Affiliation(s)
- Sudhakar Reddy Baddam
- University of Massachusetts Chan Medical School, RNA Therapeutics Institute,Worcester,MA 01655, United States of America
| | - Sudhakar Kalagara
- Department of Chemistry and Biochemistry,University of the Texas at El Paso, 500 W University Ave,El Paso,TX 79968, United States of America
| | - Krishna Kuna
- Department of Chemistry,University College of Science, Saifabad, Osmania University, Hyderabad,Telangana,India
| | - Sreenivas Enaganti
- Department of Bioinformatics, Averinbiotech Laboratories,208, 2nd Floor, Windsor Plaza, Nallakunta, Hyderabad, Telangana,India
| |
Collapse
|
6
|
Qureshi SA, Hussain L, Ibrar U, Alabdulkreem E, Nour MK, Alqahtani MS, Nafie FM, Mohamed A, Mohammed GP, Duong TQ. Radiogenomic classification for MGMT promoter methylation status using multi-omics fused feature space for least invasive diagnosis through mpMRI scans. Sci Rep 2023; 13:3291. [PMID: 36841898 PMCID: PMC9961309 DOI: 10.1038/s41598-023-30309-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/21/2023] [Indexed: 02/27/2023] Open
Abstract
Accurate radiogenomic classification of brain tumors is important to improve the standard of diagnosis, prognosis, and treatment planning for patients with glioblastoma. In this study, we propose a novel two-stage MGMT Promoter Methylation Prediction (MGMT-PMP) system that extracts latent features fused with radiomic features predicting the genetic subtype of glioblastoma. A novel fine-tuned deep learning architecture, namely Deep Learning Radiomic Feature Extraction (DLRFE) module, is proposed for latent feature extraction that fuses the quantitative knowledge to the spatial distribution and the size of tumorous structure through radiomic features: (GLCM, HOG, and LBP). The application of the novice rejection algorithm has been found significantly effective in selecting and isolating the negative training instances out of the original dataset. The fused feature vectors are then used for training and testing by k-NN and SVM classifiers. The 2021 RSNA Brain Tumor challenge dataset (BraTS-2021) consists of four structural mpMRIs, viz. fluid-attenuated inversion-recovery, T1-weighted, T1-weighted contrast enhancement, and T2-weighted. We evaluated the classification performance, for the very first time in published form, in terms of measures like accuracy, F1-score, and Matthews correlation coefficient. The Jackknife tenfold cross-validation was used for training and testing BraTS-2021 dataset validation. The highest classification performance is (96.84 ± 0.09)%, (96.08 ± 0.10)%, and (97.44 ± 0.14)% as accuracy, sensitivity, and specificity respectively to detect MGMT methylation status for patients suffering from glioblastoma. Deep learning feature extraction with radiogenomic features, fusing imaging phenotypes and molecular structure, using rejection algorithm has been found to perform outclass capable of detecting MGMT methylation status of glioblastoma patients. The approach relates the genomic variation with radiomic features forming a bridge between two areas of research that may prove useful for clinical treatment planning leading to better outcomes.
Collapse
Affiliation(s)
- Shahzad Ahmad Qureshi
- Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Islamabad, Pakistan.
| | - Lal Hussain
- Department of Computer Science and IT, Neelum Campus, The University of Azad Jammu and Kashmir, Muzaffarabad, Azad Kashmir, Pakistan. .,Department of Computer Science and IT, King Abdullah Campus, The University of Azad Jammu and Kashmir, Muzaffarabad, Azad Kashmir, Pakistan. .,Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 111 East 210th Street, Bronx, NY, 10467, USA.
| | - Usama Ibrar
- grid.461150.7Farooq Hospital, Lahore, Pakistan
| | - Eatedal Alabdulkreem
- grid.449346.80000 0004 0501 7602Department of Computer Sciences, College of Computer and Information Sciences, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671 Saudi Arabia
| | - Mohamed K. Nour
- grid.412832.e0000 0000 9137 6644Department of Computer Sciences, College of Computing and Information System, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Mohammed S. Alqahtani
- grid.412144.60000 0004 1790 7100Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, 61421 Saudi Arabia
| | - Faisal Mohammed Nafie
- grid.449051.d0000 0004 0441 5633Department of Computer Science, College of Science and Humanities at Alghat, Majmaah University, Al-Majmaah, 11952 Saudi Arabia
| | - Abdullah Mohamed
- grid.440865.b0000 0004 0377 3762Research Centre, Future University in Egypt, New Cairo, 11845 Egypt
| | - Gouse Pasha Mohammed
- grid.449553.a0000 0004 0441 5588Department of Computer and Self Development, Preparatory Year Deanship, Prince Sattam Bin Abdulaziz University, AlKharj, Saudi Arabia
| | - Tim Q. Duong
- grid.240283.f0000 0001 2152 0791Department of Radiology, Albert Einstein College of Medicine and Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467 USA
| |
Collapse
|
7
|
Wang H, Feng Y, Zhang Y, Wang T, Xu H, Zhi Y, Feng Y, Tian L, Yuan K. Siglec10-An immunosuppressor and negative predictor of survival prognosis in gliomas. Front Genet 2022; 13:873655. [PMID: 36468012 PMCID: PMC9709431 DOI: 10.3389/fgene.2022.873655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 10/28/2022] [Indexed: 07/24/2023] Open
Abstract
Glioma is a type of tumor occurring in the central nervous system. In recent decades, specific gene mutations and molecular aberrations have been used to conduct the glioma classification and clinical decisions. Siglec10 is a member of the sialic acid-binding immunoglobulin superfamily. In this study, we investigated the expression and functions of siglec10 in gliomas. We analyzed the siglec10 expression in glioma patients with immunohistochemical (IHC) staining and evaluated the survival prognosis. The high siglec10 expression had a shorter survival prognosis than the low siglec10 expression in patients, especially in malignant gliomas. Bioinformatic datasets, including TCGA and CGGA, validated the IHC results and discovered the expression of siglec10 was higher in the malignant subtype than a benign subtype of gliomas. So, siglec10 is associated with the poor prognosis of gliomas. Furthermore, the related mechanisms of siglec10 in gliomas were investigated by functional enrichment analysis, including GSEA, GO, and KEGG analysis. Siglec10 was correlated with inflammatory mediators, inflammatory cells, and inflammatory pathways in gliomas. Siglec10 might take part in the immune response in the tumor microenvironment to induce glioma's progression and metastasis. This study showed siglec10 was a biomarker in glioma, and it might be the potential target of glioma immunotherapy in the future.
Collapse
Affiliation(s)
- Hesong Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yanyan Feng
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yuxiang Zhang
- Department of Neurosurgery, Sanbo Brain Hospital Capital Medical University, Beijing, China
| | - Ting Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Heng Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuxing Zhi
- Qi-Huang Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Yuyin Feng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Lichun Tian
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kai Yuan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
8
|
Abstract
Gliomas are the most common adult and pediatric primary brain tumors. Molecular studies have identified features that can enhance diagnosis and provide biomarkers. IDH1/2 mutation with ATRX and TP53 mutations defines diffuse astrocytomas, whereas IDH1/2 mutations with 1p19q loss defines oligodendroglioma. Focal amplifications of receptor tyrosine kinase genes, TERT promoter mutation, and loss of chromosomes 10 and 13 with trisomy of chromosome 7 are characteristic features of glioblastoma and can be used for diagnosis. BRAF gene fusions and mutations in low-grade gliomas and histone H3 mutations in high-grade gliomas also can be used for diagnostics.
Collapse
Affiliation(s)
- Kristyn Galbraith
- Department of Pathology, NYU Langone Medical Center, 240 East 38th Street, 22nd Floor, New York, NY 10016, USA
| | - Matija Snuderl
- Department of Pathology, NYU Langone Medical Center, 240 East 38th Street, 22nd Floor, New York, NY 10016, USA.
| |
Collapse
|
9
|
Bacolod MD, Barany F. MGMT Epigenetics: The Influence of Gene Body Methylation and Other Insights Derived from Integrated Methylomic, Transcriptomic, and Chromatin Analyses in Various Cancer Types. Curr Cancer Drug Targets 2021; 21:360-374. [PMID: 33535955 DOI: 10.2174/1568009621666210203111620] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND MGMT (O6-methylguanine-DNA methyltransferase) is primarily responsible for limiting the activity of some widely used chemotherapeutic agents, including temozolomide (TMZ) and carmustine (BCNU). The gene encoding this protein is epigenetically regulated, and assessment of methylation at its promoter region is used to predict glioma patients' response to TMZ. METHODS In this report, we employed a bioinformatic approach to elucidate MGMT's epigenetic regulation. Integrated for the analysis were genome-wide methylation and transcription datasets for > 8,600 human tissue (representing 31 distinct cancer types) and 500 human cancer cell line samples. Also crucial to the interpretation of results were publicly available data from the ENCODE Project: tracks for histone modifications (via ChIP-seq) and DNase I hypersensitivity (via DNaseseq), as well as methylation and transcription data for representative cell lines (HeLa-S3, HMEC, K562). RESULTS AND DISCUSSION We were able to validate (perhaps more comprehensively) the contrasting influences of CpG methylation at promoter region and at gene body on MGMT transcription. While the MGMT promoter is populated by CpG sites whose methylation levels displayed high negative correlation (R) with MGMT mRNA counts, the gene body harbors CpG sites exhibiting high positive R values. The promoter CpG sites with very high negative R's across cancer types include cg12981137, cg12434587, and cg00618725. Among the notable gene body CpG sites (high positive R's across cancer types) are cg00198994 (Intron 1), cg04473030 (Intron 2), and cg07367735 (Intron 4). For certain cancer types, such as melanoma, gene body methylation appears to be a better predictor of MGMT transcription (compared to promoter methylation). In general, the CpG methylation v. MGMT expression R values are higher in cell lines relative to tissues. Also, these correlations are noticeably more prominent in certain cancer types such as colorectal, adrenocortical, esophageal, skin, and head and neck cancers, as well as glioblastoma. As expected, hypomethylation at the promoter region is associated with more open chromatin, and enrichment of histone marks H3K4m1, H3K4m2, H3K4m3, and H3K9ac. CONCLUSION Overall, our analysis illustrated the contrasting influence of promoter and gene body methylation on MGMT expression. These observations may help improve diagnostic assays for MGMT.
Collapse
Affiliation(s)
- Manny D Bacolod
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, United States
| | - Francis Barany
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, United States
| |
Collapse
|
10
|
Ahmed M, Bayoumi B, Abdallah S, Elserafy M. MGMT Immunohistochemical Expression in Colorectal Carcinoma and its Correlation with Tumor Progression. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.5879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Introduction: There is an urgent need to identify predictive features and markers for colorectal carcinoma (CRC) progression and treatment. This study aimed to assess O6-methylguanine DNA methyltransferase (MGMT) expression in CRC and correlate with the clinico-pathological aspects of the tumor, also to evaluate the relationship between different histopathologic parameters and tumor progression.
Material and Methods: The study was carried on 70 colectomy using formalin fixed paraffin embedded tumor tissue. Immunohistochemistry was used to detect MGMT expression, and clinico-pathologic aspects as well as Tumor budding, type of desmoplastic reaction, inflammatory lymphocytic milieu, pattern of invasive front and necrosis were assessed Then correlated with MGMT expression and tumor progression, using parametric and nonparametric statistical methods.
Results: MGMT Loss of expression was detected in 42.9% of CRC cases. MGMT expression status was significantly correlated with tumor stage and metastatic status (p<0.05), while it was not correlated with other clinic-pathologic features, (p>0.05). Desmoplastic reaction (DR), tumor budding, stromal tumor infiltrating lymphocytes (TIL-S) and necrosis were correlated with tumor stage (p<0.05). DR correlated with tumor budding (p<0.05). Both types of TIL and Crohn’s-like lymphoid reaction (CLR) showed a mutual correlation (p<0.05).
Conclusion: MGMT high expression and histopathologic parameters as DR, tumor budding, inflammatory lymphocytic milieu and necrosis could be correlated with CRC progression.
Collapse
|
11
|
Yogananda CGB, Shah BR, Nalawade SS, Murugesan GK, Yu FF, Pinho MC, Wagner BC, Mickey B, Patel TR, Fei B, Madhuranthakam AJ, Maldjian JA. MRI-Based Deep-Learning Method for Determining Glioma MGMT Promoter Methylation Status. AJNR Am J Neuroradiol 2021; 42:845-852. [PMID: 33664111 PMCID: PMC8115363 DOI: 10.3174/ajnr.a7029] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/21/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND AND PURPOSE O6-Methylguanine-DNA methyltransferase (MGMT) promoter methylation confers an improved prognosis and treatment response in gliomas. We developed a deep learning network for determining MGMT promoter methylation status using T2 weighted Images (T2WI) only. MATERIALS AND METHODS Brain MR imaging and corresponding genomic information were obtained for 247 subjects from The Cancer Imaging Archive and The Cancer Genome Atlas. One hundred sixty-three subjects had a methylated MGMT promoter. A T2WI-only network (MGMT-net) was developed to determine MGMT promoter methylation status and simultaneous single-label tumor segmentation. The network was trained using 3D-dense-UNets. Three-fold cross-validation was performed to generalize the performance of the networks. Dice scores were computed to determine tumor-segmentation accuracy. RESULTS The MGMT-net demonstrated a mean cross-validation accuracy of 94.73% across the 3 folds (95.12%, 93.98%, and 95.12%, [SD, 0.66%]) in predicting MGMT methylation status with a sensitivity and specificity of 96.31% [SD, 0.04%] and 91.66% [SD, 2.06%], respectively, and a mean area under the curve of 0.93 [SD, 0.01]. The whole tumor-segmentation mean Dice score was 0.82 [SD, 0.008]. CONCLUSIONS We demonstrate high classification accuracy in predicting MGMT promoter methylation status using only T2WI. Our network surpasses the sensitivity, specificity, and accuracy of histologic and molecular methods. This result represents an important milestone toward using MR imaging to predict prognosis and treatment response.
Collapse
Affiliation(s)
- C G B Yogananda
- From the Advanced Neuroscience Imaging Research Lab (C.G.B.Y., B.R.S., S.S.N., G.K.M., F.F.Y., M.C.P., B.C.W., A.J.M., J.A.M.), Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - B R Shah
- From the Advanced Neuroscience Imaging Research Lab (C.G.B.Y., B.R.S., S.S.N., G.K.M., F.F.Y., M.C.P., B.C.W., A.J.M., J.A.M.), Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - S S Nalawade
- From the Advanced Neuroscience Imaging Research Lab (C.G.B.Y., B.R.S., S.S.N., G.K.M., F.F.Y., M.C.P., B.C.W., A.J.M., J.A.M.), Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - G K Murugesan
- From the Advanced Neuroscience Imaging Research Lab (C.G.B.Y., B.R.S., S.S.N., G.K.M., F.F.Y., M.C.P., B.C.W., A.J.M., J.A.M.), Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - F F Yu
- From the Advanced Neuroscience Imaging Research Lab (C.G.B.Y., B.R.S., S.S.N., G.K.M., F.F.Y., M.C.P., B.C.W., A.J.M., J.A.M.), Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - M C Pinho
- From the Advanced Neuroscience Imaging Research Lab (C.G.B.Y., B.R.S., S.S.N., G.K.M., F.F.Y., M.C.P., B.C.W., A.J.M., J.A.M.), Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - B C Wagner
- From the Advanced Neuroscience Imaging Research Lab (C.G.B.Y., B.R.S., S.S.N., G.K.M., F.F.Y., M.C.P., B.C.W., A.J.M., J.A.M.), Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - B Mickey
- Department of Neurological Surgery (B.M., T.R.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - T R Patel
- Department of Neurological Surgery (B.M., T.R.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - B Fei
- Department of Bioengineering (B.F.), University of Texas at Dallas, Richardson, Texas
| | - A J Madhuranthakam
- From the Advanced Neuroscience Imaging Research Lab (C.G.B.Y., B.R.S., S.S.N., G.K.M., F.F.Y., M.C.P., B.C.W., A.J.M., J.A.M.), Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - J A Maldjian
- From the Advanced Neuroscience Imaging Research Lab (C.G.B.Y., B.R.S., S.S.N., G.K.M., F.F.Y., M.C.P., B.C.W., A.J.M., J.A.M.), Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
12
|
Methylation of MGMT promoter does not predict response to temozolomide in patients with glioblastoma in Donostia Hospital. Sci Rep 2020; 10:18445. [PMID: 33116181 PMCID: PMC7595088 DOI: 10.1038/s41598-020-75477-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/13/2020] [Indexed: 12/18/2022] Open
Abstract
O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status has been considered a prognostic factor in newly diagnosed glioblastoma (GBM). In this study, we evaluated the prognostic and predictive value of MGMT promoter methylation in patients with glioblastoma in Donostia Hospital. Surprisingly, methylation of MGMT promoter did not predict response to temozolomide in patients with glioblastoma in Donostia Hospital. Specifically, overall survival (OS) and progression-free survival (PFS) did not differ significantly by MGMT methylation status in our cohort. In contrast, both were longer in patients who received treatment, received more TMZ cycles, had a better general status and perform at least a partial resection. No association was detected between methylation of MGMT promoter and molecular markers such as ATRX, IDH, p53 and Ki67. These results indicate that MGMT methylation did not influence in patient survival in our cohort.
Collapse
|
13
|
Mansouri A, Hachem LD, Mansouri S, Nassiri F, Laperriere NJ, Xia D, Lindeman NI, Wen PY, Chakravarti A, Mehta MP, Hegi ME, Stupp R, Aldape KD, Zadeh G. MGMT promoter methylation status testing to guide therapy for glioblastoma: refining the approach based on emerging evidence and current challenges. Neuro Oncol 2020; 21:167-178. [PMID: 30189035 DOI: 10.1093/neuonc/noy132] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/11/2018] [Accepted: 08/29/2018] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor, with a universally poor prognosis. The emergence of molecular biomarkers has had a significant impact on histological typing and diagnosis, as well as predicting patient survival and response to treatment. The methylation status of the O6-methylguanine-DNA methyl-transferase (MGMT) gene promoter is one such molecular biomarker. Despite the strong evidence supporting the role of MGMT methylation status in prognostication, its routine implementation in clinical practice has been challenging. The methods and optimal cutoff definitions for MGMT status determination remain controversial. Variation in detection methods between laboratories presents a major challenge for consensus. Moreover, consideration of other clinical and genetic/epigenetic factors must also be incorporated into treatment decision making. In this review, we distill the available evidence to summarize our position on the optimal use of available assays, and propose strategies for resolving cases with equivocal methylation status and a framework for incorporating this important assay into research and clinical practice.
Collapse
Affiliation(s)
- Alireza Mansouri
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Laureen D Hachem
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Sheila Mansouri
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Farshad Nassiri
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Normand J Laperriere
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Daniel Xia
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Arnab Chakravarti
- Radiation Oncology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Miami, Florida, USA
| | - Monika E Hegi
- Department of Clinical Neurosciences, Lausanne University Hospital, Lausanne, Switzerland
| | - Roger Stupp
- Malnati Brain Tumor Institute of the Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kenneth D Aldape
- Department of Laboratory Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Gelareh Zadeh
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Zehnbauer BA. The Journal of Molecular Diagnostics: 20 Years Defining Professional Practice. J Mol Diagn 2019; 21:938-942. [PMID: 31635797 DOI: 10.1016/j.jmoldx.2019.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 01/09/2023] Open
Abstract
This editorial highlights 20 years of JMD defining professional practice.
Collapse
Affiliation(s)
- Barbara A Zehnbauer
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia (Editor-in-Chief).
| |
Collapse
|
15
|
Malmström A, Łysiak M, Kristensen BW, Hovey E, Henriksson R, Söderkvist P. Do we really know who has an MGMT methylated glioma? Results of an international survey regarding use of MGMT analyses for glioma. Neurooncol Pract 2019; 7:68-76. [PMID: 32025325 PMCID: PMC6993038 DOI: 10.1093/nop/npz039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background Glioma O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status informs clinical decision making. Worldwide different methods and cutoff levels are used, which can lead to discordant methylation results. Methods We conducted an international survey to clarify which methods are regularly used and why. We also explored opinions regarding international consensus on methods and cutoff. Results The survey had 152 respondents from 25 countries. MGMT methylation status is determined for all glioblastomas in 37% of laboratories. The most common methods are methylation-specific polymerase chain reaction (msPCR) (37%) and pyrosequencing (34%). A method is selected for simplicity (56%), cost-effectiveness (50%), and reproducibility of results (52%). For sequencing, the number of CpG sites analyzed varies from 1–3 up to more than 16. For 50% of laboratories, the company producing the kit determines which CpG sites are examined, whereas 33% select the sites themselves. Selection of cutoff is equally distributed among a cutoff defined in the literature, by the local laboratory, or by the outside laboratory performing the analysis. This cutoff varies, reported from 1% to 30%, and in 1 laboratory tumor is determined as methylated in case of 1 methylated CpG site of 17 analyzed. Some report tumors as unmethylated or weakly vs highly methylated. An international consensus on MGMT methylation method and cutoff is warranted by 66% and 76% of respondents, respectively. The method preferred would be msPCR (45%) or pyrosequencing (42%), whereas 18% suggest next-generation sequencing. Conclusion Although analysis of MGMT methylation status is routine, there is controversy regarding laboratory methods and cutoff level. Most respondents favor development of international consensus guidelines.
Collapse
Affiliation(s)
- Annika Malmström
- Department of Advanced Home Care, Linköping University, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Sweden
| | - Małgorzata Łysiak
- Department of Clinical and Experimental Medicine, Linköping University, Sweden
| | - Bjarne Winther Kristensen
- Department of Pathology, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark
| | - Elizabeth Hovey
- Department of Medical Oncology, Nelune Comprehensive Cancer Centre, Prince of Wales Hospital, Randwick, Sydney, NSW, Australia.,University of New South Wales, Sydney, Australia
| | | | | |
Collapse
|
16
|
Jovanović N, Mitrović T, Cvetković VJ, Tošić S, Vitorović J, Stamenković S, Nikolov V, Kostić A, Vidović N, Krstić M, Jevtović-Stoimenov T, Pavlović D. The Impact of MGMT Promoter Methylation and Temozolomide Treatment in Serbian Patients with Primary Glioblastoma. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E34. [PMID: 30717206 PMCID: PMC6409652 DOI: 10.3390/medicina55020034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 01/29/2019] [Indexed: 01/25/2023]
Abstract
Background and objective: Despite recent advances in treatment, glioblastoma (GBM) remains the most lethal and aggressive brain tumor. A continuous search for a reliable molecular marker establishes the methylation status of the O6-methylguanine-DNA methyltransferase (MGMT) gene promoter as a key prognostic factor in primary glioblastoma. The aim of our study was to screen Serbian patients with primary glioblastoma for an MGMT promoter hypermethylation and to evaluate its associations with overall survival (OS) and sensitivity to temozolomide (TMZ) treatment. Materials and methods: A cohort of 30 Serbian primary glioblastoma patients treated with radiation therapy and chemotherapy were analyzed for MGMT promoter methylation and correlated with clinical data. Results: MGMT methylation status was determined in 25 out of 30 primary glioblastomas by methylation-specific PCR (MSP). MGMT promoter hypermethylation was detected in 12 out of 25 patients (48%). The level of MGMT promoter methylation did not correlate with patients' gender (p = 0.409), age (p = 0.536), and OS (p = 0.394). Treatment with TMZ significantly prolonged the median survival of a patient (from 5 to 15 months; p < 0.001). Conclusions: Due to a small cohort of primary GBM patients, our study is not sufficient for definitive conclusions regarding the prognostic value of MGMT methylation for the Serbian population. Our preliminary data suggest a lack of association between MGMT promoter methylation and overall survival and a significant correlation of TMZ treatment with overall survival. Further population-based studies are needed to assess the prognostic value of the MGMT promoter methylation status for patients with primary glioblastoma.
Collapse
Affiliation(s)
- Nikola Jovanović
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Tatjana Mitrović
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Vladimir J Cvetković
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Svetlana Tošić
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Jelena Vitorović
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Slaviša Stamenković
- University of Niš, Department of Biology and Ecology, Faculty of Sciences and Mathematics, 18000 Niš, Serbia.
| | - Vesna Nikolov
- University of Niš, Faculty of Medicine, Clinic of Neurosurgery, Clinical Center, 18000 Niš, Serbia.
| | - Aleksandar Kostić
- University of Niš, Faculty of Medicine, Clinic of Neurosurgery, Clinical Center, 18000 Niš, Serbia.
| | - Nataša Vidović
- University of Niš, Faculty of Medicine, Pathology and Pathological Anatomy Center, 18000 Niš, Serbia.
| | - Miljan Krstić
- University of Niš, Faculty of Medicine, Pathology and Pathological Anatomy Center, 18000 Niš, Serbia.
| | | | - Dušica Pavlović
- University of Niš, Faculty of Medicine, Institute of Biochemistry, 18000 Niš, Serbia.
| |
Collapse
|
17
|
Abstract
Immunohistochemistry (IHC) can be applied to diagnostic aspects of pathologic examination to provide aid in assignment of lineage and histologic type of cancer. Increasingly, however, IHC is widely used to provide prognostic and predictive (theranostic) information about the neoplastic disease. A refinement of theranostic application of IHC can be seen in the use of "genomic probing" where antibody staining results are directly correlated with an underlying genetic alteration in the tumor (somatic mutations) and/or the patient (germline constitution). All these aspects of IHC find their best use in guiding the oncologists in the optimal use of therapy for the patients.
Collapse
Affiliation(s)
| | | | - Semir Vranić
- College of Medicine, Qatar University, Doha, Qatar
| | | |
Collapse
|
18
|
|
19
|
Panagopoulos I, Gorunova L, Leske H, Niehusmann P, Johannessen LE, Staurseth J, Øino N, Meling TR, Heim S, Micci F, Brandal P. Pyrosequencing Analysis of MGMT Promoter Methylation in Meningioma. Cancer Genomics Proteomics 2018; 15:379-385. [PMID: 30194078 DOI: 10.21873/cgp.20096] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/28/2018] [Accepted: 07/08/2018] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Methylation of the O6-methylguanine-DNA methyltransferase (MGMT) gene promoter is a well-established predictor of response to the DNA-alkylating agent temozolomide in patients with glioblastoma. MATERIALS AND METHODS Pyrosequencing analysis was used to determine the MGMT promoter methylation status in 61 meningiomas, to clarify whether it might have a predictive role. RESULTS Only two tumors (3%) had a mean methylation frequency higher than the cut-off value of 10% for the four CpG sites examined. CONCLUSION The methylation of the MGMT promoter is uncommon, or occurs at a low frequency in meningiomas. There is no convincing rationale to test such tumors for their MGMT methylation status in a clinical setting.
Collapse
Affiliation(s)
- Ioannis Panagopoulos
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ludmila Gorunova
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Henning Leske
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Pitt Niehusmann
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Lene E Johannessen
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Julie Staurseth
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Nina Øino
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Torstein R Meling
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Neurosurgery, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Sverre Heim
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Francesca Micci
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Petter Brandal
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
20
|
Dovek L, Nguyen NT, Ozer BH, Li N, Elashoff RM, Green RM, Liau L, Nghiemphu PL, Cloughesy TF, Lai A. Correlation of commercially available quantitative MGMT (O-6-methylguanine-DNA methyltransferase) promoter methylation scores and GBM patient survival. Neurooncol Pract 2018; 6:194-202. [PMID: 31386024 DOI: 10.1093/nop/npy028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/01/2018] [Indexed: 11/13/2022] Open
Abstract
Background Between 2011 and 2016, O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation testing at University of California Los Angeles (UCLA) was performed through LabCorp, using a threshold of 2 to distinguish MGMT methylated from unmethylated tumors. In this study, we sought to determine whether the magnitude of the methylation score correlated with outcome. Methods We identified 165 newly diagnosed glioblastoma (GBM) isocitrate dehydrogenase (IDH) wild-type and temozolomide-treated upfront patients at UCLA and Kaiser Permanente Los Angeles with LabCorp-derived quantitative MGMT scores obtained on pretreatment tissue samples. Using LabCorp's threshold, we found 102 unmethylated and 63 methylated patients. We then further substratified each group based on the magnitude of the score, and performed Kaplan-Meier and Cox regression analyses of overall survival (OS) and progression-free survival (PFS). Results We validated that the standard LabCorp threshold of 2 could separate our cohort by survival, showing longer OS and PFS for MGMT methylated patients vs unmethylated patients. Cox regression analysis confirmed that MGMT (<1) patients had worse outcome, with OS and PFS hazard ratios of 2.375 (P = .053) and 2.463 (P = .023), respectively, when compared to the MGMT (1-1.99) patients. Contrary to our expectation, when we substratified the ≥2 (methylated) group, we did not find a dose-dependent relationship between the magnitude of MGMT methylation and improved survival. Conclusions The MGMT unmethylated group contains a partially methylated group (greater than 1) that shares survival benefits similar to the methylated group. However, we did not demonstrate an association of very high methylation scores with increased survival. These findings will require validation in additional independent clinical data sets.
Collapse
Affiliation(s)
- Laura Dovek
- University of California Los Angeles (UCLA) Department of Neurology, Los Angeles
| | - Nhung T Nguyen
- University of California Los Angeles (UCLA) Department of Neurology, Los Angeles
| | - Byram H Ozer
- University of California Los Angeles (UCLA) Department of Neurology, Los Angeles
| | - Ning Li
- UCLA Department of Biomathematics, Los Angeles
| | | | | | - Linda Liau
- UCLA Department of Neurosurgery, Los Angeles
| | - P Leia Nghiemphu
- University of California Los Angeles (UCLA) Department of Neurology, Los Angeles
| | - Timothy F Cloughesy
- University of California Los Angeles (UCLA) Department of Neurology, Los Angeles
| | - Albert Lai
- University of California Los Angeles (UCLA) Department of Neurology, Los Angeles
| |
Collapse
|
21
|
Romano FJ, Guadagno E, Solari D, Borrelli G, Pignatiello S, Cappabianca P, Del Basso De Caro M. ATM and p53 combined analysis predicts survival in glioblastoma multiforme patients: A clinicopathologic study. J Cell Biochem 2018; 119:4867-4877. [PMID: 29369420 DOI: 10.1002/jcb.26699] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/23/2018] [Indexed: 12/19/2022]
Abstract
Glioblastoma is one of the most malignant cancers, with a distinguishing dismal prognosis: surgery followed by chemo- and radiotherapy represents the current standard of care, and chemo- and radioresistance underlie disease recurrence and short overall survival of patients suffering from this malignancy. ATM is a kinase activated by autophosphorylation upon DNA doublestrand breaks arising from errors during replication, byproducts of metabolism, chemotherapy or ionizing radiations; TP53 is one of the most popular tumor suppressor, with a preeminent role in DNA damage response and repair. To study the effects of the immunohistochemical expression of p-ATM and p53 in glioblastoma patients, 21 cases were retrospectively examined. In normal brain tissue, p-ATM was expressed only in neurons; conversely, in tumors cells, the protein showed a variable cytoplasmic expression (score: +,++,+++), with being completely undetectable in three cases. Statistical analysis revealed that high p-ATM score (++/+++) strongly correlated to shorter survival (P = 0.022). No difference in overall survival was registered between p53 normally expressed (NE) and overexpressed (OE) glioblastoma patients (P = 0.669). Survival analysis performed on the results from combined assessment of the two proteins showed that patients with NE p53 /low pATM score had longer overall survival than the NE p53/ high pATM score counterpart. Cox-regression analysis confirmed this finding (HR = 0.025; CI 95% = 0.002-0.284; P = 0.003). Our study outlined the immunohistochemical expression of p-ATM/p53 in glioblastomas and provided data on their possible prognostic/predictive of response role. A "non-oncogene addiction" to ATM for NEp53 glioblastoma could be postulated, strengthening the rationale for development of ATM inhibiting drugs.
Collapse
Affiliation(s)
| | - Elia Guadagno
- Department of Advanced Biomedical Sciences, Pathology Section, Division of Neurosurgery - University of Naples Federico II, Naples, Italy
| | - Domenico Solari
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Division of Neurosurgery - University of Naples Federico II, Naples, Italy
| | - Giorgio Borrelli
- Department of Advanced Biomedical Sciences, Pathology Section, Division of Neurosurgery - University of Naples Federico II, Naples, Italy
| | - Sara Pignatiello
- Department of Advanced Biomedical Sciences, Pathology Section, Division of Neurosurgery - University of Naples Federico II, Naples, Italy
| | - Paolo Cappabianca
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, Division of Neurosurgery - University of Naples Federico II, Naples, Italy
| | - Marialaura Del Basso De Caro
- Department of Advanced Biomedical Sciences, Pathology Section, Division of Neurosurgery - University of Naples Federico II, Naples, Italy
| |
Collapse
|
22
|
Wang PF, Liu N, Song HW, Yao K, Jiang T, Li SW, Yan CX. IDH-1R132H mutation status in diffuse glioma patients: implications for classification. Oncotarget 2017; 7:31393-400. [PMID: 27120786 PMCID: PMC5058765 DOI: 10.18632/oncotarget.8918] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 04/11/2016] [Indexed: 01/03/2023] Open
Abstract
WHO2007 grading of diffuse gliomas in adults is well-established. However, IDH mutations make classification of gliomas according to the WHO2007 edition controversial. Here, we characterized IDH-1R132H mut status in a cohort of 670 adult patients with different WHO2007 grades of diffuse glioma. Patient characteristics, clinical data and prognoses were obtained from medical records. Patients with IDH-1R132H mut were younger and had better clinical outcomes than those without mutations. Differences in age among patients with astrocytomas of different WHO2007 grades were eliminated after patients were grouped based on IDH-1R132H status. IDH-1R132H mut was present more often in patients with lower Ki-67 and MGMT protein levels and higher mutant p53 levels. Ki-67 was also strongly associated with WHO2007 grade independently of IDH-1R132H mut status. Moreover, patients with Ki-67<30 survived longer than those with Ki-67≥30, regardless of IDH-1R132H mut status. Patients in the IDH-1R132H mut group with lower MGMT protein levels also had better clinical outcomes than those in other groups. Our results indicate that to better treat gliomas, IDH mutation status should be included when determining WHO2007 grade in glioma patients.
Collapse
Affiliation(s)
- Peng-Fei Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Ning Liu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Hong-Wang Song
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Kun Yao
- Department of Pathology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Neurosurgical Institute, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Shou-Wei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chang-Xiang Yan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Wang L, Li Z, Liu C, Chen L, Liu L, Hu Z, Zhao L, Lu D, Teng L. Comparative assessment of three methods to analyze MGMT methylation status in a series of 350 gliomas and gangliogliomas. Pathol Res Pract 2017; 213:1489-1493. [PMID: 29103769 DOI: 10.1016/j.prp.2017.10.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/29/2017] [Accepted: 10/07/2017] [Indexed: 11/27/2022]
Abstract
MGMT promoter methylation is considered as a prognostic and predictive biomarker indicating response to chemotherapy and radiotherapy in glioblastoma. A number of different methods and platforms including pyrosequencing (PSQ), quantitative methylation-specific PCR (qMSP) and immunohistochemistry (IHC), methylation-sensitive high resolution melting (MS-HRM) and NGS (Next Generation Sequencing) have been used to detect MGMT promoter methylation in gliomas. However, controversy remains about the most appropriate method to use for analyzing MGMT status. The MGMT promoter methylation status of a total of 350 gliomas and gangliogliomas was examined using PSQ, qMSP and IHC in parallel. Using PSQ as a recommended standard method, the sensitivity, specificity, positive/negative predictive value and correlation with the other assays were calculated. Among 350 glioma and ganglioglioma cases, the MGMT promoter tested positive for methylation in 53.1%, 55.4%, and 70.3% of the cases by PSQ, qMSP and IHC, respectively. The sensitivity and specificity of qMSP were 97.8% and 92.7%, respectively. Twelve cases that tested positive for methylation using qMSP were negative according to PSQ, and four cases that were negative according to qMSP tested positive according to PSQ. The concordance rate between PSQ and qMSP was 90.8%. The sensitivity and specificity of IHC for the detection of MGMT at the protein level were 84.4% and 45.7%, respectively. The concordance rate between PSQ and IHC was 30.8%. This study demonstrated that qMSP is an effective and rapid detection method for routine use in pathology laboratories for the identification of MGMT promoter methylation. A combination of IHC and qMSP assays can provide high sensitivity and specificity for the prediction of MGMT status. A few cases that tested negative with PSQ did harbor MGMT promoter methylation, as confirmed by qMSP and sequencing, and this subgroup of patients may benefit from temozolomide.
Collapse
Affiliation(s)
- Leiming Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Zhuo Li
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Cuicui Liu
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Li Chen
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Li Liu
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Zeliang Hu
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lihong Zhao
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Dehong Lu
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lianghong Teng
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
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
|
Abstract
Melanoma is a malignant tumor of melanocytes and is considered to be the most aggressive cancer among all skin diseases. The pathogenesis of melanoma has not been well documented, which may restrict the research and development of biomarkers and therapies. To date, several genetic and epigenetic factors have been identified as contributing to the development and progression of melanoma. Besides the findings on genetic susceptibilities, the recent progress in epigenetic studies has revealed that loss of the DNA hydroxymethylation mark, 5-hydroxymethylcytosine (5-hmC), along with high levels of DNA methylation at promoter regions of several tumor suppressor genes in melanoma, may serve as biomarkers for melanoma. Moreover, 5-Aza-2′-deoxycytidine, an epigenetic modifier causing DNA demethylation, and ten-eleven translocation family dioxygenase (TET), which catalyzes the generation of 5-hmC, demonstrate therapeutic potential in melanoma treatment. In this review, we will summarize the latest progress in research on DNA methylation/hydroxymethylation in melanoma, and we will discuss and provide insight for epigenetic biomarkers and therapies for melanoma. Particularly, we will discuss the role of DNA hydroxymethylation in melanoma infiltrating immune cells, which may also serve as a potential target for melanoma treatment.
Collapse
|
26
|
Zhang J, Liu H, Tong H, Wang S, Yang Y, Liu G, Zhang W. Clinical Applications of Contrast-Enhanced Perfusion MRI Techniques in Gliomas: Recent Advances and Current Challenges. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:7064120. [PMID: 29097933 PMCID: PMC5612612 DOI: 10.1155/2017/7064120] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/23/2017] [Indexed: 01/12/2023]
Abstract
Gliomas possess complex and heterogeneous vasculatures with abnormal hemodynamics. Despite considerable advances in diagnostic and therapeutic techniques for improving tumor management and patient care in recent years, the prognosis of malignant gliomas remains dismal. Perfusion-weighted magnetic resonance imaging techniques that could noninvasively provide superior information on vascular functionality have attracted much attention for evaluating brain tumors. However, nonconsensus imaging protocols and postprocessing analysis among different institutions impede their integration into standard-of-care imaging in clinic. And there have been very few studies providing a comprehensive evidence-based and systematic summary. This review first outlines the status of glioma theranostics and tumor-associated vascular pathology and then presents an overview of the principles of dynamic contrast-enhanced MRI (DCE-MRI) and dynamic susceptibility contrast-MRI (DSC-MRI), with emphasis on their recent clinical applications in gliomas including tumor grading, identification of molecular characteristics, differentiation of glioma from other brain tumors, treatment response assessment, and predicting prognosis. Current challenges and future perspectives are also highlighted.
Collapse
Affiliation(s)
- Junfeng Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Heng Liu
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Haipeng Tong
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Sumei Wang
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Yizeng Yang
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Weiguo Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Chongqing Clinical Research Center for Imaging and Nuclear Medicine, Chongqing 400042, China
| |
Collapse
|
27
|
Kaul KL, Sabatini LM, Tsongalis GJ, Caliendo AM, Olsen RJ, Ashwood ER, Bale S, Benirschke R, Carlow D, Funke BH, Grody WW, Hayden RT, Hegde M, Lyon E, Murata K, Pessin M, Press RD, Thomson RB. The Case for Laboratory Developed Procedures: Quality and Positive Impact on Patient Care. Acad Pathol 2017; 4:2374289517708309. [PMID: 28815200 PMCID: PMC5528950 DOI: 10.1177/2374289517708309] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/06/2017] [Accepted: 04/10/2017] [Indexed: 12/16/2022] Open
Abstract
An explosion of knowledge and technology is revolutionizing medicine and patient care. Novel testing must be brought to the clinic with safety and accuracy, but also in a timely and cost-effective manner, so that patients can benefit and laboratories can offer testing consistent with current guidelines. Under the oversight provided by the Clinical Laboratory Improvement Amendments, laboratories have been able to develop and optimize laboratory procedures for use in-house. Quality improvement programs, interlaboratory comparisons, and the ability of laboratories to adjust assays as needed to improve results, utilize new sample types, or incorporate new mutations, information, or technologies are positive aspects of Clinical Laboratory Improvement Amendments oversight of laboratory-developed procedures. Laboratories have a long history of successful service to patients operating under Clinical Laboratory Improvement Amendments. A series of detailed clinical examples illustrating the quality and positive impact of laboratory-developed procedures on patient care is provided. These examples also demonstrate how Clinical Laboratory Improvement Amendments oversight ensures accurate, reliable, and reproducible testing in clinical laboratories.
Collapse
Affiliation(s)
- Karen L. Kaul
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Evanston, IL, USA
| | - Linda M. Sabatini
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Evanston, IL, USA
| | - Gregory J. Tsongalis
- Laboratory for Clinical Genomics and Advanced Technology, Department of Pathology, Dartmouth Hitchcock Medical Center and Norris Cotton Cancer Center, Lebanon, NH, USA
- Laboratory Medicine, Dartmouth Hitchcock Medical Center and Norris Cotton Cancer Center, Lebanon, NH, USA
| | - Angela M. Caliendo
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - Randall J. Olsen
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | | | | | - Robert Benirschke
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Evanston, IL, USA
| | - Dean Carlow
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Birgit H. Funke
- Laboratory for Molecular Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Wayne W. Grody
- Departments of Pathology and Laboratory Medicine, Pediatrics and Human Genetics, UCLA School of Medicine, Los Angeles, CA, USA
| | - Randall T. Hayden
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Madhuri Hegde
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Elaine Lyon
- Pathology Department, University of Utah School of Medicine/ARUP Laboratories, Salt Lake City, UT, USA
| | - Kazunori Murata
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melissa Pessin
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard D. Press
- Department of Pathology and Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Richard B. Thomson
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Evanston, IL, USA
| |
Collapse
|
28
|
Xia D, Reardon DA, Bruce JL, Lindeman NI. The Clinical Implications of Inconsistently Methylated Results from Glioblastoma MGMT Testing by Replicate Methylation-Specific PCR. J Mol Diagn 2016; 18:864-871. [PMID: 27639200 DOI: 10.1016/j.jmoldx.2016.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 05/15/2016] [Accepted: 06/20/2016] [Indexed: 11/25/2022] Open
Abstract
The methylation status of the promoter of the O6-methylguanine DNA methyltransferase gene (MGMT) is an established prognostic and predictive biomarker of glioblastoma (GBM). At the Center for Advanced Molecular Diagnostics, MGMT testing is performed by methylation-specific PCR with multiple replicates, leading to three types of reportable results: methylated, unmethylated, and inconsistently methylated. An inconsistently methylated result is reported when a methylated peak is seen in some but not all of the PCR replicates from a single DNA sample. To better understand the clinical implications of these results, we performed a retrospective review of all MGMT testing at our laboratory over a 5-year period, and correlated test results with outcome and specimen-quality data. This review yielded several novel findings. First, inconsistent MGMT methylation on replicate methylation-specific PCR is not uncommon, composes 12% (58/465) of our GBM results. Second, inconsistently methylated GBM cases are associated with relatively poor overall survival (more similar to unmethylated than to methylated cases). Third and interestingly, there appears to be a dose-response relationship between patient survival and the extent of methylation in inconsistently methylated GBMs. Finally, our analyses of specimen-quality data suggest that a combination of technical factors (eg, small samples) and tumor biology may explain inconsistent MGMT results on replicate methylation-specific PCR testing.
Collapse
Affiliation(s)
- Daniel Xia
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jacqueline L Bruce
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Neal I Lindeman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
29
|
Ashby LS, Smith KA, Stea B. Gliadel wafer implantation combined with standard radiotherapy and concurrent followed by adjuvant temozolomide for treatment of newly diagnosed high-grade glioma: a systematic literature review. World J Surg Oncol 2016; 14:225. [PMID: 27557526 PMCID: PMC4997737 DOI: 10.1186/s12957-016-0975-5] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 08/09/2016] [Indexed: 12/04/2022] Open
Abstract
Since 2003, only two chemotherapeutic agents, evaluated in phase III trials, have been approved by the US Food and Drug Administration for treatment of newly diagnosed high-grade glioma (HGG): Gliadel wafers (intracranially implanted local chemotherapy) and temozolomide (TMZ) (systemic chemotherapy). Neither agent is curative, but each has been shown to improve median overall survival (OS) compared to radiotherapy (RT) alone. To date, no phase III trial has tested these agents when used in sequential combination; however, a number of smaller trials have reported favorable results. We performed a systematic literature review to evaluate the combination of Gliadel wafers with standard RT (60 Gy) plus concurrent and adjuvant TMZ (RT/TMZ) for newly diagnosed HGG. A literature search was conducted for the period of January 1995 to September 2015. Data were extracted and categorized, and means and ranges were determined. A total of 11 publications met criteria, three prospective trials and eight retrospective studies, representing 411 patients who received Gliadel plus standard RT/TMZ. Patients were similar in age, gender, and performance status. The weighted mean of median OS was 18.2 months (ten trials, n = 379, range 12.7 to 21.3 months), and the weighted mean of median progression-free survival was 9.7 months (seven trials, n = 287, range 7 to 12.9 months). The most commonly reported grade 3 and 4 adverse events were myelosuppression (10.22 %), neurologic deficit (7.8 %), and healing abnormalities (4.3 %). Adverse events reflected the distinct independent safety profiles of Gliadel wafers and RT/TMZ, with little evidence of enhanced toxicity from their use in sequential combination. In the 11 identified trials, an increased benefit from sequentially combining Gliadel wafers with RT/TMZ was strongly suggested. Median OS tended to be improved by 3 to 4 months beyond that observed for Gliadel wafers or TMZ when used alone in the respective phase III trials. Larger prospective trials of Gliadel plus RT/TMZ are warranted.
Collapse
Affiliation(s)
- Lynn S Ashby
- Department of Neurology, Barrow Neurological Institute, 500 W. Thomas Rd, Suite 300, Phoenix, AZ, 85013, USA.
| | - Kris A Smith
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, 85013, USA
| | - Baldassarre Stea
- Department of Radiation Oncology, Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
| |
Collapse
|
30
|
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: 7] [Impact Index Per Article: 0.8] [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
|
31
|
Localization of specialized intestinal metaplasia and the molecular alterations in Barrett esophagus in a Japanese population: an analysis of biopsy samples based on the "Seattle" biopsy protocol. Hum Pathol 2016; 51:32-40. [PMID: 27067780 DOI: 10.1016/j.humpath.2015.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/11/2015] [Accepted: 12/16/2015] [Indexed: 02/07/2023]
Abstract
It remains unclear why Barrett esophagus (BE)-associated adenocarcinoma (EAC) frequently occurs in the 0 to 3 o'clock area of the BE. The aims of this study were to clarify the localization of specialized intestinal metaplasia (SIM) as a precancerous lesion and of molecular alterations among different locations using 4-quadrant biopsies based on the "Seattle" protocol. We prospectively evaluated microsatellite instability; methylation status at the APC, CDKN2A, hMLH1, RUNX3, and MGMT genes; the immunoreactivity of the monoclonal antibody Das-1 for the colonic phenotype; and Ki-67 staining in 10 early EACs and 128 biopsy samples from 32 BE patients. Among the molecular changes, only APC gene hypermethylation was an independent predictive marker of EAC (odds ratio, 24.4; P = .01). SIM was more frequently identified in the 0 to 3 o'clock quadrant than in the 6 to 9 o'clock quadrant (P = .08). The Ki-67 index was higher in SIM than in the columnar-lined epithelium (CLE) without goblet cells (P < .0001) and in both SIM and CLE with Das-1 reactivity than in those without (P = .04 and P = .06, respectively). Furthermore, the index was relatively higher in the 0 to 3 o'clock quadrant than in the 6 to 9 o'clock quadrant in cases with Das-1 reactivity. RUNX3 methylation was more frequently found in SIM than in CLE (P = .04), whereas the incidence of the other biomarkers did not show a significant difference between the 0 to 3 o'clock and 6 to 9 o'clock areas, nor between SIM and CLE. SIM with Das-1 reactivity, but not molecular alterations, in the 0 to 3 o'clock quadrant may have higher proliferative activity compared to the other areas of the BE.
Collapse
|
32
|
Abstract
Sorting and grading of glial tumors by the WHO classification provide clinicians with guidance as to the predicted course of the disease and choice of treatment. Nonetheless, histologically identical tumors may have very different outcome and response to treatment. Molecular markers that carry both diagnostic and prognostic information add useful tools to traditional classification by redefining tumor subtypes within each WHO category. Therefore, molecular markers have become an integral part of tumor assessment in modern neuro-oncology and biomarker status now guides clinical decisions in some subtypes of gliomas. The routine assessment of IDH status improves histological diagnostic accuracy by differentiating diffuse glioma from reactive gliosis. It carries a favorable prognostic implication for all glial tumors and it is predictive for chemotherapeutic response in anaplastic oligodendrogliomas with codeletion of 1p/19q chromosomes. Glial tumors that contain chromosomal codeletion of 1p/19q are defined as tumors of oligodendroglial lineage and have favorable prognosis. MGMT promoter methylation is a favorable prognostic marker in astrocytic high-grade gliomas and it is predictive for chemotherapeutic response in anaplastic gliomas with wild-type IDH1/2 and in glioblastoma of the elderly. The clinical implication of other molecular markers of gliomas like mutations of EGFR and ATRX genes and BRAF fusion or point mutation is highlighted. The potential of molecular biomarker-based classification to guide future therapeutic approach is discussed and accentuated.
Collapse
|
33
|
MGMT testing allows for personalised therapy in the temozolomide era. Tumour Biol 2015; 37:87-96. [PMID: 26518768 DOI: 10.1007/s13277-015-4240-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022] Open
Abstract
Adjuvant temozolomide (TMZ)-based chemoradiation is the standard of care for most glioblastoma patients (GBMs); however, a large proportion of these patients do not respond to TMZ. Silencing of the O(6)-methylguanine-DNA methyltransferase (MGMT) promoter is thought to induce chemosensitivity, and testing for methylation may allow for patient stratification; however, this has yet to become routine clinical practice despite an abundance of literature on the subject. The databases PubMed, Embase, The Cochrane Library, Science Direct and Medline were searched for relevant articles published between 1999 and 2015. Articles utilising MGMT testing in glioblastomas, and treatment of glioblastomas with temozolomide were assessed. Immunohistochemistry, methylation-specific PCR (MSP), reverse transcriptase PCR, pyrosequencing and bisulphite sequencing were the main testing methods identified. Nested-MSP techniques produced poor correlation with survival, whilst bisulphite sequencing showed no evident benefit over MSP. Testing is limited by sample quality and contamination; however, efforts are made to minimise this. Strong evidence for MGMT-based personalised therapy was presented in the elderly but remains controversial in the entire GBM population. MGMT testing presents many obstacles yet to be overcome, and these warrant attention prior to the routine implementation of MGMT testing to aid decision making in GBMs. However, there is evidence to support its use, particularly in the elderly.
Collapse
|
34
|
Tuominen R, Jewell R, van den Oord JJ, Wolter P, Stierner U, Lindholm C, Hertzman Johansson C, Lindén D, Johansson H, Frostvik Stolt M, Walker C, Snowden H, Newton-Bishop J, Hansson J, Egyházi Brage S. MGMT promoter methylation is associated with temozolomide response and prolonged progression-free survival in disseminated cutaneous melanoma. Int J Cancer 2015; 136:2844-53. [PMID: 25400033 DOI: 10.1002/ijc.29332] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 10/16/2014] [Indexed: 11/11/2022]
Abstract
To investigate the predictive and prognostic value of O(6) -methylguanine DNA methyltransferase (MGMT) inactivation by analyses of promoter methylation in pretreatment tumor biopsies from patients with cutaneous melanoma treated with dacarbazine (DTIC) or temozolomide (TMZ) were performed. The patient cohorts consisted of Belgian and Swedish disseminated melanoma patients. Patients were subdivided into those receiving single-agent treatment with DTIC/TMZ (cohort S, n = 74) and those treated with combination chemotherapy including DTIC/TMZ (cohort C, n = 79). Median follow-up was 248 and 336 days for cohort S and cohort C, respectively. MGMT promoter methylation was assessed by three methods. The methylation-related transcriptional silencing of MGMT mRNA expression was assessed by real-time RT-PCR. Response to chemotherapy and progression-free survival (PFS) and overall survival were correlated to MGMT promoter methylation status. MGMT promoter methylation was detected in tumor biopsies from 21.5 % of the patients. MGMT mRNA was found to be significantly lower in tumors positive for MGMT promoter methylation compared to tumors without methylation in both treatment cohorts (p < 0.005). DTIC/TMZ therapy response rate was found to be significantly associated with MGMT promoter methylation in cohort S (p = 0.0005), but did not reach significance in cohort C (p = 0.16). Significantly longer PFS was observed among patients with MGMT promoter-methylated tumors (p = 0.002). Multivariate Cox regression analysis identified presence of MGMT promoter methylation as an independent variable associated with longer PFS. Together, this implies that MGMT promoter methylation is associated with response to single-agent DTIC/TMZ and longer PFS in disseminated cutaneous melanoma.
Collapse
Affiliation(s)
- Rainer Tuominen
- Department of Oncology and Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
DNA Methylation Levels of Melanoma Risk Genes Are Associated with Clinical Characteristics of Melanoma Patients. BIOMED RESEARCH INTERNATIONAL 2015; 2015:376423. [PMID: 26106605 PMCID: PMC4461735 DOI: 10.1155/2015/376423] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/23/2015] [Indexed: 12/17/2022]
Abstract
In melanoma development, oncogenic process is mediated by genetic and epigenetic mutations, and few studies have so far explored the role of DNA methylation either as predisposition factor or biomarker. We tested patient samples for germline CDKN2A methylation status and found no evidence of inactivation by promoter hypermethylation. We have also investigated the association of clinical characteristics of samples with the DNA methylation pattern of twelve genes relevant for melanomagenesis. Five genes (BAP1, MGMT, MITF, PALB2, and POT1) presented statistical association between blood DNA methylation levels and either CDKN2A-mutation status, number of lesions, or Breslow thickness. In tumors, five genes (KIT, MGMT, MITF, TERT, and TNF) exhibited methylation levels significantly different between tumor groups including acral compared to nonacral melanomas and matched primary lesions and metastases. Our data pinpoint that the methylation level of eight melanoma-associated genes could potentially represent markers for this disease both in peripheral blood and in tumor samples.
Collapse
|
36
|
Nymoen DA, Holth A, Hetland Falkenthal TE, Tropé CG, Davidson B. CIAPIN1 and ABCA13 are markers of poor survival in metastatic ovarian serous carcinoma. Mol Cancer 2015; 14:44. [PMID: 25889687 PMCID: PMC4336750 DOI: 10.1186/s12943-015-0317-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/09/2015] [Indexed: 02/07/2023] Open
Abstract
Background The objective of this study was to investigate the expression and clinical role of 14 genes previously shown to be associated with chemotherapy response and/or progression-free survival in a smaller series of ovarian serous carcinoma effusions. Methods Advanced-stage serous ovarian carcinoma effusions (n = 150) were analyzed for mRNA expression of AKR1C1, ABCA4, ABCA13, ABCB10, BIRC6, CASP9, CIAPIN1, FAS, MGMT, MUTYH, POLH, SRC, TBRKB and XPA using quantitative real-time PCR. mRNA expression was studied for association with clinicopathologic parameters, including chemotherapy response and survival. Results ABCA4 mRNA expression was significantly related to better (complete) chemotherapy response at diagnosis in the entire cohort (p = 0.018), whereas higher POLH mRNA levels were significantly related to better chemoresponse at diagnosis in analysis to 58 patients with pre-chemotherapy effusions treated with standard chemotherapy (carboplatin + paclitaxel; p = 0.023). In univariate survival analysis for patients with pre-chemotherapy effusions (n = 77), CIAPIN1 mRNA expression was significantly related to shorter overall (p = 0.007) and progression-free (p = 0.038) survival, whereas ABCA13 mRNA expression was significantly related to shorter OS (p = 0.024). Higher CIAPIN1 mRNA expression was an independent marker of poor overall survival in Cox multivariate analysis (p = 0.044). Conclusions Our data identify ABCA4 and POLH as markers of better chemotherapy response in metastatic serous carcinoma. CIAPIN1 and ABCA13 may be novel markers of poor outcome in pre-chemotherapy serous carcinoma effusions.
Collapse
Affiliation(s)
- Dag Andre Nymoen
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, Montebello, N-0310, Oslo, Norway.
| | - Arild Holth
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, Montebello, N-0310, Oslo, Norway.
| | | | - Claes G Tropé
- Department of Gynecologic Oncology, Oslo University Hospital, Norwegian Radium Hospital, N-0310, Oslo, Norway. .,University of Oslo, Faculty of Medicine, Institute of Clinical Medicine, N-0316, Oslo, Norway.
| | - Ben Davidson
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, Montebello, N-0310, Oslo, Norway. .,University of Oslo, Faculty of Medicine, Institute of Clinical Medicine, N-0316, Oslo, Norway.
| |
Collapse
|
37
|
Majchrzak-Celińska A, Paluszczak J, Szalata M, Barciszewska AM, Nowak S, Kleszcz R, Sherba A, Baer-Dubowska W. The methylation of a panel of genes differentiates low-grade from high-grade gliomas. Tumour Biol 2015; 36:3831-41. [PMID: 25563195 DOI: 10.1007/s13277-014-3025-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/26/2014] [Indexed: 12/29/2022] Open
Abstract
Epigenetic changes play an important role in the pathogenesis of gliomas and have the potential to become clinically useful biomarkers. The aim of this study was the evaluation of the profile of promoter methylation of 13 genes selected based on their anticipated diagnostic and/or prognostic value. Methylation-specific PCR (MSP) was used to assess the methylation status of MGMT, ERCC1, hMLH1, ATM, CDKN2B (p15INK4B), p14ARF, CDKN2A (p16INK4A), RASSF1A, RUNX3, GATA6, NDRG2, PTEN, and RARβ in a subset of 95 gliomas of different grades. Additionally, the methylation status of MGMT and NDRG2 was analyzed using pyrosequencing (PSQ). The results revealed that the methylation index of individual glioma patients correlates with World Health Organization (WHO) tumor grade and patient's age. RASSF1A, RUNX3, GATA6, and MGMT were most frequently methylated, whereas the INK4B-ARF-INK4A locus, PTEN, RARβ, and ATM were methylated to a lesser extent. ERCC1, hMLH1, and NDRG2 were unmethylated. RUNX3 methylation correlated with WHO tumor grade and patient's age. PSQ confirmed significantly higher methylation levels of MGMT and NDRG2 as compared with normal, non-cancerous brain tissue. To conclude, DNA methylation of a whole panel of selected genes can serve as a tool for glioma aggressiveness prediction.
Collapse
Affiliation(s)
- Aleksandra Majchrzak-Celińska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, ul. Święcickiego 4, 60-781, Poznań, Poland
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Siegal T. Clinical impact of molecular biomarkers in gliomas. J Clin Neurosci 2014; 22:437-44. [PMID: 25533211 DOI: 10.1016/j.jocn.2014.10.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 10/01/2014] [Indexed: 12/21/2022]
Abstract
The World Health Organization (WHO) classification system for glial tumors provides guidance as to the predicted course of the disease and choice of treatment. However, histologically identical tumors may have a very different outcome and response to treatment. Molecular markers that carry both diagnostic and prognostic information add valuable tools by redefining tumor subtypes within each WHO category. Therefore, molecular biomarkers have become an integral part of tumor assessment in modern neuro-oncology and biomarker status now guides clinical decisions in some subtypes of gliomas, including anaplastic oligodendroglioma and glioblastoma in the elderly. This review discusses the prognostic and predictive impact of molecular markers that have undergone extensive study in recent years. The clinical relevance of contemporary molecular classification of gliomas using the routine assessment of IDH mutations, promoter methylation of MGMT, chromosomal deletion of 1p/19q, mutations of EGFR and ATRX genes, and BRAF fusion or point mutation is highlighted. The potential of molecular biomarker-based classification to guide future therapeutic approach is discussed and accentuated.
Collapse
Affiliation(s)
- Tali Siegal
- Center for Neuro-Oncology, Davidoff Institute of Oncology, Rabin Medical Center, Campus Beilinson, 49100 Petach Tikva, Israel.
| |
Collapse
|
39
|
Bajpai M, Das KM, Lefferts J, Lisovsky M, Mashimo H, Phillips WA, Srivastava A, To H. Molecular epidemiology of and genetic susceptibility to esophageal cancer. Ann N Y Acad Sci 2014; 1325:40-8. [PMID: 25266013 DOI: 10.1111/nyas.12517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The following, from the 12th OESO World Conference: Cancers of the Esophagus, includes commentaries on clonal evolution in Barrett's carcinogenesis; biomarkers for early detection of esophageal cancer; the role of the methylguanine methyl transferase biomarker in the management of adenocarcinoma; and the discovery of high-risk genes in families.
Collapse
Affiliation(s)
- Manisha Bajpai
- Division of GI/Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Yong RL, Wu T, Mihatov N, Shen MJ, Brown MA, Zaghloul KA, Park GE, Park JK. Residual tumor volume and patient survival following reoperation for recurrent glioblastoma. J Neurosurg 2014; 121:802-9. [PMID: 25061868 DOI: 10.3171/2014.6.jns132038] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Maximal safe tumor resection is part of the standard of care for patients with newly diagnosed glioblastoma. The role of reoperation in the care of patients with recurrent glioblastoma is less clear, and less than a quarter of patients undergo a second surgery. Previous studies have identified preoperative variables associated with the improved survival of patients following reoperation, and guidelines for the selection of patients for reoperation have been devised and validated. In this study, the authors analyzed the relative survival benefit of maximal safe tumor removal in a series of patients with recurrent glioblastoma who all underwent reoperation. METHODS In this longitudinal study, the clinical and radiological data of 97 consecutive patients who underwent reoperation for recurrent glioblastoma were prospectively collected. Multiple regression analyses and Kaplan-Meier plotting were performed to identify pre- and postoperative clinical and radiological variables associated with increased survival following reoperation. RESULTS The median postoperative survival of all patients following reoperation was 12.4 months (95% confidence interval [CI] 9.0-15.6 months). Multiple Cox regression analysis revealed that patients with large (> 3 cm(3)) residual tumors following reoperation had significantly decreased survival relative to those with residual tumors that were small (> 0-3 cm(3); hazard ratio [HR] = 3.10, 95% CI 1.69-5.70; p < 0.001) or radiologically absent (0 cm(3); HR = 5.82, 95% CI 2.98-11.37; p < 0.001). Large residual tumors had faster rates of subsequent regrowth than small (odds ratio [OR] = 4.22, 95% CI 1.19-14.97; p = 0.026) or radiologically absent (OR = 11.00, 95% CI 2.79-43.43; p = 0.001) residual tumors, and a faster regrowth rate was significantly associated with decreased survival (HR = 4.01, 95% CI 2.26-7.14; p < 0.001). CONCLUSIONS The overall survival of patients with recurrent glioblastoma who underwent reoperations increased with decreasing postoperative residual tumor volumes. For patients meeting prognostic criteria for reoperation, the surgical goal should be to minimize residual tumor volume to maximize overall survival. Clinical trial registration no.: NCT00060541 ( ClinicalTrials.gov ).
Collapse
Affiliation(s)
- Raymund L Yong
- National Institute of Neurological Disorders and Stroke and
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Toffolatti L, Scquizzato E, Cavallin S, Canal F, Scarpa M, Stefani PM, Gherlinzoni F, Dei Tos AP. MGMT promoter methylation and correlation with protein expression in primary central nervous system lymphoma. Virchows Arch 2014; 465:579-86. [PMID: 25031012 DOI: 10.1007/s00428-014-1622-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 06/04/2014] [Accepted: 06/30/2014] [Indexed: 12/26/2022]
Abstract
The O (6)-methylguanine-DNA-methyltransferase (MGMT) gene encodes for a DNA repairing enzyme of which silencing by promoter methylation is involved in brain tumorigenesis. MGMT promoter methylation represents a favorable prognostic factor and has been associated with a better response to alkylating agents in glioma and systemic lymphoma. Primary central nervous system lymphoma (PCNSL) is a rare and aggressive extranodal malignant lymphoma. The current standard of care, based on high-dose methotrexate chemotherapy, has improved prognosis but outcome remains poor for a majority of patients. Therapeutic progress in this field is conditioned by limited biological and molecular knowledge about the disease. Temozolomide has recently emerged as an alternative option for PCNSL treatment. We aimed to analyze the MGMT gene methylation status in a series of 24 PCNSLs, to investigate the relationship between methylation status of the gene and immunohistochemical expression of MGMT protein and to evaluate the possible prognostic significance of these biomarkers. Our results confirm that methylation of the MGMT gene and loss of MGMT protein are frequent events in these lymphomas (54 % of our cases) and suggest that they are gender and age related. MGMT methylation showed high correlation with loss of protein expression (concordance correlation coefficient = -0.49; Fisher exact test: p < 0.01), different from what has been observed in other brain tumors. In the subgroup of ten patients who received high dose chemotherapy, the presence of methylated MGMT promoter (n = 4), seems to be associated with a prolonged overall survival (>60 months in three of four patients). The prognostic significance of these molecular markers in PCNSL needs to be further studied in groups of patients treated in a homogeneous way.
Collapse
Affiliation(s)
- L Toffolatti
- Department of Pathology, Treviso General Hospital, Piazzale Ospedale 1, 31100, Treviso, TV, Italy,
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Nagel ZD, Chaim IA, Samson LD. Inter-individual variation in DNA repair capacity: a need for multi-pathway functional assays to promote translational DNA repair research. DNA Repair (Amst) 2014; 19:199-213. [PMID: 24780560 DOI: 10.1016/j.dnarep.2014.03.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Why does a constant barrage of DNA damage lead to disease in some individuals, while others remain healthy? This article surveys current work addressing the implications of inter-individual variation in DNA repair capacity for human health, and discusses the status of DNA repair assays as potential clinical tools for personalized prevention or treatment of disease. In particular, we highlight research showing that there are significant inter-individual variations in DNA repair capacity (DRC), and that measuring these differences provides important biological insight regarding disease susceptibility and cancer treatment efficacy. We emphasize work showing that it is important to measure repair capacity in multiple pathways, and that functional assays are required to fill a gap left by genome wide association studies, global gene expression and proteomics. Finally, we discuss research that will be needed to overcome barriers that currently limit the use of DNA repair assays in the clinic.
Collapse
Affiliation(s)
- Zachary D Nagel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Isaac A Chaim
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leona D Samson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
43
|
Fumagalli C, Della Pasqua S, Bagnardi V, Cardillo A, Sporchia A, Colleoni M, Viale G, Barberis M, Pruneri G. Prevalence and clinicopathologic correlates of O⁶-methylguanine-DNA methyltransferase methylation status in patients with triple-negative breast cancer treated preoperatively by alkylating drugs. Clin Breast Cancer 2014; 14:285-90. [PMID: 24709436 DOI: 10.1016/j.clbc.2014.02.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 02/14/2014] [Accepted: 02/24/2014] [Indexed: 11/17/2022]
Abstract
BACKGROUND Predictive factors of benefit from specific chemotherapy regimens are not currently available in triple-negative breast cancer (TNBC). MGMT (O(6)-methylguanine-DNA methyltransferase) controls DNA repair pathways, and its epigenetic silencing is used for predicting the response to the alkylating drug temozolomide in patients with glioma. MATERIALS AND METHODS The study population was composed of 84 patients with TNBC treated with alkylating agents and evaluated for clinicopathologic parameters (tumor shrinkage and pathologic complete response [pCR]). MGMT methylation status was assessed in formalin-fixed, paraffin-embedded tumor specimens by pyrosequencing. The samples were categorized as methylated (mean methylation value > 5%), indeterminate (4%-5%), and unmethylated (≤ 3%). RESULTS MGMT methylation status was successfully evaluated in all the cases: 58.3% were methylated; 27.4%, unmethylated; and 14.3%, indeterminate. MGMT methylation was observed in 80%, 62%, and 29% of patients showing a 100%, 99% to 30%, and < 30% tumor reduction, respectively, a trend not achieving statistical significance (P = .23). There was no association between MGMT methylation status and pCR. CONCLUSION The present study provided evidence that pyrosequencing performs well for the evaluation of MGMT methylation even in small bioptic samples, suggesting that it could be reliably used in translational studies of preoperative clinical trials. Although there was an association trend between high methylation levels and clinical response to therapy, no statistically significant association with the pCR was found. Further studies in larger series of patients are warranted for ascertaining the putative clinical role of MGMT in patients with TNBC.
Collapse
Affiliation(s)
| | | | - Vincenzo Bagnardi
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - Anna Cardillo
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| | - Andrea Sporchia
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| | - Marco Colleoni
- Division of Medical Senology, European Institute of Oncology, Milan, Italy
| | - Giuseppe Viale
- Division of Pathology, European Institute of Oncology, Milan, Italy; University of Milan School of Medicine; Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Massimo Barberis
- Division of Pathology, European Institute of Oncology, Milan, Italy
| | - Giancarlo Pruneri
- Division of Pathology, European Institute of Oncology, Milan, Italy; University of Milan School of Medicine; Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy.
| |
Collapse
|
44
|
Minoo P. Toward a Molecular Classification of Colorectal Cancer: The Role of MGMT. Front Oncol 2013; 3:266. [PMID: 24151575 PMCID: PMC3798865 DOI: 10.3389/fonc.2013.00266] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/04/2013] [Indexed: 11/23/2022] Open
Abstract
O6-methylguanine DNA methyltransferase (MGMT) is a DNA repair enzyme with the ability to protect cells from DNA mutations by removing alkyl groups from the O6 position of guanine. Colon mucosa is exposed to the direct effects of environmental carcinogens and therefore maintaining a proficient DNA repair system is very important to stay protected against DNA mutagenesis. Loss of MGMT expression is almost exclusively associated with methylation of CpG islands in the MGMT gene promoter region which is found in approximately 40% of colorectal cancers. The role of MGMT loss in colorectal tumorigenesis is complex but numerous studies have documented methylation of this gene even in the normal appearing mucosa as well as in aberrant crypt foci, suggesting that MGMT methylation can be regarded as an early event or “field defect” in colon cancer neoplasia. The focus of this perspective is the role of MGMT in different pathways of colorectal carcinogenesis as well as the implication of this molecule in treatment decisions in colorectal cancer patients.
Collapse
Affiliation(s)
- Parham Minoo
- Calgary Laboratory Services, Department of Pathology, University of Calgary , Calgary, AB , Canada
| |
Collapse
|