1
|
Lu ZG, Shen J, Yang J, Wang JW, Zhao RC, Zhang TL, Guo J, Zhang X. Nucleic acid drug vectors for diagnosis and treatment of brain diseases. Signal Transduct Target Ther 2023; 8:39. [PMID: 36650130 PMCID: PMC9844208 DOI: 10.1038/s41392-022-01298-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/08/2022] [Accepted: 12/21/2022] [Indexed: 01/18/2023] Open
Abstract
Nucleic acid drugs have the advantages of rich target selection, simple in design, good and enduring effect. They have been demonstrated to have irreplaceable superiority in brain disease treatment, while vectors are a decisive factor in therapeutic efficacy. Strict physiological barriers, such as degradation and clearance in circulation, blood-brain barrier, cellular uptake, endosome/lysosome barriers, release, obstruct the delivery of nucleic acid drugs to the brain by the vectors. Nucleic acid drugs against a single target are inefficient in treating brain diseases of complex pathogenesis. Differences between individual patients lead to severe uncertainties in brain disease treatment with nucleic acid drugs. In this Review, we briefly summarize the classification of nucleic acid drugs. Next, we discuss physiological barriers during drug delivery and universal coping strategies and introduce the application methods of these universal strategies to nucleic acid drug vectors. Subsequently, we explore nucleic acid drug-based multidrug regimens for the combination treatment of brain diseases and the construction of the corresponding vectors. In the following, we address the feasibility of patient stratification and personalized therapy through diagnostic information from medical imaging and the manner of introducing contrast agents into vectors. Finally, we take a perspective on the future feasibility and remaining challenges of vector-based integrated diagnosis and gene therapy for brain diseases.
Collapse
Affiliation(s)
- Zhi-Guo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| | - Jie Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Jun Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Jing-Wen Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Rui-Chen Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Tian-Lu Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Jing Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| |
Collapse
|
2
|
Montella L, Cuomo M, Del Gaudio N, Buonaiuto M, Costabile D, Visconti R, Di Risi T, Vinciguerra R, Trio F, Ferraro S, Bove G, Facchini G, Altucci L, Chiariotti L, Della Monica R. Epigenetic alterations in glioblastomas: Diagnostic, prognostic and therapeutic relevance. Int J Cancer 2022. [PMID: 36479695 DOI: 10.1002/ijc.34381] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/17/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022]
Abstract
Glioblastoma, the most common and heterogeneous tumor affecting brain parenchyma, is dismally characterized by a very poor prognosis. Thus, the search of new, more effective treatments is a vital need. Here, we will review the druggable epigenetic features of glioblastomas that are, indeed, currently explored in preclinical studies and in clinical trials for the development of more effective, personalized treatments. In detail, we will review the studies that have led to the identification of epigenetic signatures, IDH mutations, MGMT gene methylation, histone modification alterations, H3K27 mutations and epitranscriptome landscapes of glioblastomas, in each case discussing the corresponding targeted therapies and their potential efficacy. Finally, we will emphasize how recent technological improvements permit to routinely investigate many glioblastoma epigenetic biomarkers in clinical practice, further enforcing the hope that personalized drugs, targeting specific epigenetic features, could be in future a therapeutic option for selected patients.
Collapse
Affiliation(s)
- Liliana Montella
- ASL NA2 NORD, Oncology Operative Unit, "Santa Maria delle Grazie" Hospital, Pozzuoli, Italy
| | - Mariella Cuomo
- CEINGE Biotecnologie Avanzate scarl, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Nunzio Del Gaudio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Michela Buonaiuto
- CEINGE Biotecnologie Avanzate scarl, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Davide Costabile
- CEINGE Biotecnologie Avanzate scarl, Naples, Italy.,SEMM-European School of Molecular Medicine, University of Naples "Federico II", Naples, Italy
| | - Roberta Visconti
- CEINGE Biotecnologie Avanzate scarl, Naples, Italy.,Institute for the Experimental Endocrinology and Oncology, Italian National Council of Research, Naples, Italy
| | - Teodolinda Di Risi
- CEINGE Biotecnologie Avanzate scarl, Naples, Italy.,Department of Public Health, University of Naples "Federico II", Naples, Italy
| | | | | | - Sara Ferraro
- CEINGE Biotecnologie Avanzate scarl, Naples, Italy
| | - Guglielmo Bove
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gaetano Facchini
- ASL NA2 NORD, Oncology Operative Unit, "Santa Maria delle Grazie" Hospital, Pozzuoli, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.,BIOGEM, Ariano Irpino, Italy
| | - Lorenzo Chiariotti
- CEINGE Biotecnologie Avanzate scarl, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Rosa Della Monica
- CEINGE Biotecnologie Avanzate scarl, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
3
|
Liu D, Cheng Y, Qiao S, Liu M, Ji Q, Zhang BL, Mei QB, Zhou S. Nano-Codelivery of Temozolomide and siPD-L1 to Reprogram the Drug-Resistant and Immunosuppressive Microenvironment in Orthotopic Glioblastoma. ACS NANO 2022; 16:7409-7427. [PMID: 35549164 DOI: 10.1021/acsnano.1c09794] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Glioblastoma (GBM) is an invasive cancer with high mortality in central nervous system. Resistance to temozolomide (TMZ) and immunosuppressive microenvironment lead to low outcome of the standardized treatment for GBM. In this study, a 2-deoxy-d-glucose modified lipid polymer nanoparticle loaded with TMZ and siPD-L1 (TMZ/siPD-L1@GLPN/dsb) was prepared to reprogram the TMZ-resistant and immunosuppressive microenvironment in orthotopic GBM. TMZ/siPD-L1@GLPN/dsb simultaneously delivered a large amount of TMZ and siPD-L1 to the deep area of the orthotopic TMZ-resistant GBM tissue. By inhibiting PD-L1 protein expression, TMZ/siPD-L1@GLPN/dsb markedly augmented the percentage of CD3+CD8+IFN-γ+ cells (Teff cells) and reduced the percentage of CD4+CD25+FoxP3+ cells (Treg cells) in orthotopic TMZ-resistant GBM tissue, which enhanced T-cell mediated cytotoxicity on orthotopic TMZ-resistant GBM. Moreover, TMZ/siPD-L1@GLPN/dsb obviously augmented the sensitivity of orthotopic TMZ-resistant GBM to TMZ through decreasing the protein expression of O6-methyl-guanine-DNA methyltransferase (MGMT) in TMZ-resistant GBM cells. Thus, TMZ/siPD-L1@GLPN/dsb markedly restrained the growth of orthotopic TMZ-resistant GBM and extended the survival time of orthotopic GBM rats through reversing a TMZ-resistant and immunosuppressive microenvironment. TMZ/siPD-L1@GLPN/dsb shows potential application to treat orthotopic TMZ-resistant GBM.
Collapse
Affiliation(s)
- Daozhou Liu
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Ying Cheng
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Sai Qiao
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Miao Liu
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Qifeng Ji
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Bang-Le Zhang
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Qi-Bing Mei
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Siyuan Zhou
- Department of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
4
|
McAleenan A, Kelly C, Spiga F, Kernohan A, Cheng HY, Dawson S, Schmidt L, Robinson T, Brandner S, Faulkner CL, Wragg C, Jefferies S, Howell A, Vale L, Higgins JPT, Kurian KM. Prognostic value of test(s) for O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation for predicting overall survival in people with glioblastoma treated with temozolomide. Cochrane Database Syst Rev 2021; 3:CD013316. [PMID: 33710615 PMCID: PMC8078495 DOI: 10.1002/14651858.cd013316.pub2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Glioblastoma is an aggressive form of brain cancer. Approximately five in 100 people with glioblastoma survive for five years past diagnosis. Glioblastomas that have a particular modification to their DNA (called methylation) in a particular region (the O6-methylguanine-DNA methyltransferase (MGMT) promoter) respond better to treatment with chemotherapy using a drug called temozolomide. OBJECTIVES To determine which method for assessing MGMT methylation status best predicts overall survival in people diagnosed with glioblastoma who are treated with temozolomide. SEARCH METHODS We searched MEDLINE, Embase, BIOSIS, Web of Science Conference Proceedings Citation Index to December 2018, and examined reference lists. For economic evaluation studies, we additionally searched NHS Economic Evaluation Database (EED) up to December 2014. SELECTION CRITERIA Eligible studies were longitudinal (cohort) studies of adults with diagnosed glioblastoma treated with temozolomide with/without radiotherapy/surgery. Studies had to have related MGMT status in tumour tissue (assessed by one or more method) with overall survival and presented results as hazard ratios or with sufficient information (e.g. Kaplan-Meier curves) for us to estimate hazard ratios. We focused mainly on studies comparing two or more methods, and listed brief details of articles that examined a single method of measuring MGMT promoter methylation. We also sought economic evaluations conducted alongside trials, modelling studies and cost analysis. DATA COLLECTION AND ANALYSIS Two review authors independently undertook all steps of the identification and data extraction process for multiple-method studies. We assessed risk of bias and applicability using our own modified and extended version of the QUality In Prognosis Studies (QUIPS) tool. We compared different techniques, exact promoter regions (5'-cytosine-phosphate-guanine-3' (CpG) sites) and thresholds for interpretation within studies by examining hazard ratios. We performed meta-analyses for comparisons of the three most commonly examined methods (immunohistochemistry (IHC), methylation-specific polymerase chain reaction (MSP) and pyrosequencing (PSQ)), with ratios of hazard ratios (RHR), using an imputed value of the correlation between results based on the same individuals. MAIN RESULTS We included 32 independent cohorts involving 3474 people that compared two or more methods. We found evidence that MSP (CpG sites 76 to 80 and 84 to 87) is more prognostic than IHC for MGMT protein at varying thresholds (RHR 1.31, 95% confidence interval (CI) 1.01 to 1.71). We also found evidence that PSQ is more prognostic than IHC for MGMT protein at various thresholds (RHR 1.36, 95% CI 1.01 to 1.84). The data suggest that PSQ (mainly at CpG sites 74 to 78, using various thresholds) is slightly more prognostic than MSP at sites 76 to 80 and 84 to 87 (RHR 1.14, 95% CI 0.87 to 1.48). Many variants of PSQ have been compared, although we did not see any strong and consistent messages from the results. Targeting multiple CpG sites is likely to be more prognostic than targeting just one. In addition, we identified and summarised 190 articles describing a single method for measuring MGMT promoter methylation status. AUTHORS' CONCLUSIONS PSQ and MSP appear more prognostic for overall survival than IHC. Strong evidence is not available to draw conclusions with confidence about the best CpG sites or thresholds for quantitative methods. MSP has been studied mainly for CpG sites 76 to 80 and 84 to 87 and PSQ at CpG sites ranging from 72 to 95. A threshold of 9% for CpG sites 74 to 78 performed better than higher thresholds of 28% or 29% in two of three good-quality studies making such comparisons.
Collapse
Affiliation(s)
- Alexandra McAleenan
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Claire Kelly
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Francesca Spiga
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ashleigh Kernohan
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Hung-Yuan Cheng
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Sarah Dawson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Applied Research Collaboration West (ARC West) , University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Lena Schmidt
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tomos Robinson
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Claire L Faulkner
- Bristol Genetics Laboratory, Pathology Sciences, Southmead Hospital, Bristol, UK
| | - Christopher Wragg
- Bristol Genetics Laboratory, Pathology Sciences, Southmead Hospital, Bristol, UK
| | - Sarah Jefferies
- Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| | - Amy Howell
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Luke Vale
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Julian P T Higgins
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Applied Research Collaboration West (ARC West) , University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
| | - Kathreena M Kurian
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol Medical School: Brain Tumour Research Centre, Public Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
5
|
Mikkelsen VE, Dai HY, Stensjøen AL, Berntsen EM, Salvesen Ø, Solheim O, Torp SH. MGMT Promoter Methylation Status Is Not Related to Histological or Radiological Features in IDH Wild-type Glioblastomas. J Neuropathol Exp Neurol 2021; 79:855-862. [PMID: 32688383 DOI: 10.1093/jnen/nlaa060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/25/2020] [Accepted: 06/03/2020] [Indexed: 11/15/2022] Open
Abstract
O6-methylguanine DNA methyltransferase (MGMT) promoter methylation is an important favorable predictive marker in patients with glioblastoma (GBM). We hypothesized that MGMT status could be a surrogate marker of pretreatment tumor biology observed as histopathological and radiological features. Apart from some radiological studies aiming to noninvasively predict the MGMT status, few studies have investigated relationships between MGMT status and phenotypical tumor biology. We have therefore aimed to investigate such relationships in 85 isocitrate dehydrogenase (IDH) wild-type GBMs. MGMT status was determined by methylation-specific PCR and was assessed for associations with 22 histopathological features, immunohistochemical proliferative index and microvessel density measurements, conventional magnetic resonance imaging characteristics, preoperative speed of tumor growth, and overall survival. None of the investigated histological or radiological features were significantly associated with MGMT status. Methylated MGMT status was a significant independent predictor of improved overall survival. In conclusion, our results suggest that MGMT status is not related to the pretreatment phenotypical biology in IDH wild-type GBMs. Furthermore, our findings suggest the survival benefit of MGMT methylated GBMs is not due to an inherently less aggressive tumor biology, and that conventional magnetic resonance imaging features cannot be used to noninvasively predict the MGMT status.
Collapse
Affiliation(s)
- Vilde Elisabeth Mikkelsen
- From the Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology
| | - Hong Yan Dai
- Department of Pathology, St Olav's University Hospital
| | - Anne Line Stensjøen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology
| | - Erik Magnus Berntsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology.,Department of Radiology and Nuclear Medicine, St. Olav's University Hospital
| | | | - Ole Solheim
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology.,Department of Neurosurgery, St. Olav's University Hospital, Trondheim, Norway
| | - Sverre Helge Torp
- From the Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology.,Department of Pathology, St Olav's University Hospital
| |
Collapse
|
6
|
Ness C, Katta K, Garred Ø, Kumar T, Olstad OK, Petrovski G, Moe MC, Noer A. Integrated differential DNA methylation and gene expression of formalin-fixed paraffin-embedded uveal melanoma specimens identifies genes associated with early metastasis and poor prognosis. Exp Eye Res 2020; 203:108426. [PMID: 33387485 DOI: 10.1016/j.exer.2020.108426] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 12/09/2020] [Accepted: 12/22/2020] [Indexed: 12/23/2022]
Abstract
PURPOSE Uveal melanoma (UM) is an aggressive malignancy, in which nearly 50% of the patients die from metastatic disease. Aberrant DNA methylation is recognized as an important epigenomic event in carcinogenesis. Formalin-fixed paraffin-embedded (FFPE) samples represent a valuable source of tumor tissue, and recent technology has enabled the use of these samples in genome-wide DNA methylation analyses. Our aim was to investigate differential DNA methylation in relation to histopathological classification and survival data. In addition we sought to identify aberrant DNA methylation of genes that could be associated with metastatic disease and poor survival. METHODS FFPE samples from UM patients (n = 23) who underwent enucleation of the eye in the period 1976-1989 were included. DNA methylation was assessed using the Illumina Infinium HumanMethylation450 array and coupled to histopathological data, Cancer Registry of Norway- (registered UM metastasis) and Norwegian Cause of Death Registry- (time and cause of death) data. Differential DNA methylation patterns contrasting histological classification, survival data and clustering properties were investigated. Survival groups were defined as "Early metastasis" (metastases and death within 2-5 years after enucleation, n = 8), "Late metastasis" (metastases and death within 9-21 years after enucleation, n = 7) and "No metastasis" (no detected metastases ≥18 years after enucleation, n = 8). A subset of samples were selected based on preliminary multi-dimensional scaling (MDS) plots, histopathological classification, chromosome 3 status, survival status and clustering properties; "Subset Early metastasis" (n = 4) vs "Subset No metastasis" (n = 4). Bioinformatics analyses were conducted in the R statistical software. Differentially methylated positions (DMPs) and differentially methylated regions (DMRs) in various comparisons were assessed. Gene expression of relevant subgroups was determined by microarray analysis and quantitative reverse-transcription polymerase chain reaction (qRT-PCR). RESULTS DNA methylation analyses identified 2 clusters that separated the samples according to chromosome 3 status. Cluster 1 consisted of samples (n = 5) with chromosome 3 disomy (D3), while Cluster 2 was comprised of samples (n = 15) with chromosome 3 monosomy (M3). 1212 DMRs and 9386 DMPs were identified in M3 vs D3. No clear clusters were formed based on our predefined survival groups ("Early", "Late", "No") nor histopathological classification (Epithelioid, Mixed, Spindle). We identified significant changes in DNA methylation (beta FC ≥ 0.2, adjusted p < 0.05) between two sample subsets (n = 8). "Subset Early metastasis" (n = 4) vs "Subset No metastasis" (n = 4) identified 348 DMPs and 36 DMRs, and their differential gene expression by microarray showed that 14 DMPs and 2 DMRs corresponded to changes in gene expression (FC ≥ 1.5, p < 0.05). RNF13, ZNF217 and HYAL1 were hypermethylated and downregulated in "Subset Early metastasis" vs "Subset No metastasis" and could be potential tumor suppressors. TMEM200C, RGS10, ADAM12 and PAM were hypomethylated and upregulated in "Subset Early metastasis vs "Subset No metastasis" and could be potential oncogenes and thus markers of early metastasis and poor prognosis in UM. CONCLUSIONS DNA methylation profiling showed differential clustering of samples according to chromosome 3 status: Cluster 1 (D3) and Cluster 2 (M3). Integrated differential DNA methylation and gene expression of two subsets of samples identified genes associated with early metastasis and poor prognosis. RNF13, ZNF217 and HYAL1 are hypermethylated and candidate tumor suppressors, while TMEM200C, RGS10, ADAM12 and PAM are hypomethylated and candidate oncogenes linked to early metastasis. UM FFPE samples represent a valuable source for methylome studies and enable long-time follow-up.
Collapse
Affiliation(s)
- Charlotte Ness
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kirankumar Katta
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, Norway
| | - Theresa Kumar
- Department of Pathology, Oslo University Hospital, Norway
| | | | - Goran Petrovski
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Morten C Moe
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Agate Noer
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
7
|
Tanaka S, Akimoto J, Narita Y. Determination of the cutoff point of the absolute value of MGMTmRNA for predicting the therapeutic resistance to temozolomide in glioblastoma. J Neurosurg Sci 2020; 64:434-439. [PMID: 33236861 DOI: 10.23736/s0390-5616.17.04209-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND We previously reported that the absolute value of O<sup>6</sup>-methylguanine-DNA methyltransferase (MGMT) messenger RNA (mRNA) obtained using real-time reverse transcription polymerase chain reaction (RT-PCR) might be useful for predicting both the prognosis and the results of therapy for glioblastoma (GB) treated by temozolomide (TMZ). METHODS MGMT mRNA was measured in 55 newly diagnosed cases of GB less than 75 and had a Karnofsky performance status (KPS) of at least 60 by real-time reverse transcription polymerase chain reaction (RT-PCR) using the TaqMan probe. A receiver operating characteristic analysis was performed to determine the cutoff points for progression free survival (PFS) and overall survival (OS). RESULTS In 55 patients with GB, 1200 and 3600 for PFS, 1200, 2100 and 2900 copies/μgRNA for OS were the candidate cutoff points. Significantly longer PFS and OS were observed in patients who did not exceed 1200 copies/μg RNA. CONCLUSIONS One thousand and two hundred copies/μg RNA appeared to be the most reasonable cutoff point of MGMTmRNA in GB for deciding to use other anti-tumor drugs such as Bevacizumab together with TMZ.
Collapse
Affiliation(s)
- Satoshi Tanaka
- Department of Neuro-Oncology and Neurosurgery, Tokyo Nishi Tokushukai Hospital, Akishima, Japan -
| | - Jiro Akimoto
- Department of Neurosurgery, Tokyo Medical University Hospital, Tokyo, Japan
| | - Yoshitaka Narita
- Division of Neurosurgery, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
8
|
Lu Y, Patel M, Natarajan K, Ughratdar I, Sanghera P, Jena R, Watts C, Sawlani V. Machine learning-based radiomic, clinical and semantic feature analysis for predicting overall survival and MGMT promoter methylation status in patients with glioblastoma. Magn Reson Imaging 2020; 74:161-170. [PMID: 32980505 DOI: 10.1016/j.mri.2020.09.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/27/2020] [Accepted: 09/08/2020] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Survival varies in patients with glioblastoma due to intratumoral heterogeneity and radiomics/imaging biomarkers have potential to demonstrate heterogeneity. The objective was to combine radiomic, semantic and clinical features to improve prediction of overall survival (OS) and O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status from pre-operative MRI in patients with glioblastoma. METHODS A retrospective study of 181 MRI studies (mean age 58 ± 13 years, mean OS 497 ± 354 days) performed in patients with histopathology-proven glioblastoma. Tumour mass, contrast-enhancement and necrosis were segmented from volumetric contrast-enhanced T1-weighted imaging (CE-T1WI). 333 radiomic features were extracted and 16 Visually Accessible Rembrandt Images (VASARI) features were evaluated by two experienced neuroradiologists. Top radiomic, VASARI and clinical features were used to build machine learning models to predict MGMT status, and all features including MGMT status were used to build Cox proportional hazards regression (Cox) and random survival forest (RSF) models for OS prediction. RESULTS The optimal cut-off value for MGMT promoter methylation index was 12.75%; 42 radiomic features exhibited significant differences between high and low-methylation groups. However, model performance accuracy combining radiomic, VASARI and clinical features for MGMT status prediction varied between 45 and 67%. For OS predication, the RSF model based on clinical, VASARI and CE radiomic features achieved the best performance with an average iAUC of 96.2 ± 1.7 and C-index of 90.0 ± 0.3. CONCLUSIONS VASARI features in combination with clinical and radiomic features from the enhancing tumour show promise for predicting OS with a high accuracy in patients with glioblastoma from pre-operative volumetric CE-T1WI.
Collapse
Affiliation(s)
- Yiping Lu
- Neuroradiology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Mindelsohn Way, Edgbaston, Birmingham B15 2TH, UK; Radiology, Huashan Hospital, Fudan University, Wulumuqi Middle Road, Shanghai, China
| | - Markand Patel
- Neuroradiology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Mindelsohn Way, Edgbaston, Birmingham B15 2TH, UK; University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Kal Natarajan
- Medical Physics, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Mindelsohn Way, Edgbaston, Birmingham B15 2TH, UK
| | - Ismail Ughratdar
- Neurosurgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Mindelsohn Way, Edgbaston, Birmingham B15 2TH, UK
| | - Paul Sanghera
- Clinical Oncology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Mindelsohn Way, Edgbaston, Birmingham B15 2TH, UK
| | - Raj Jena
- Oncology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK
| | - Colin Watts
- University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Neurosurgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Mindelsohn Way, Edgbaston, Birmingham B15 2TH, UK
| | - Vijay Sawlani
- Neuroradiology, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Mindelsohn Way, Edgbaston, Birmingham B15 2TH, UK; University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
9
|
Butler M, Pongor L, Su YT, Xi L, Raffeld M, Quezado M, Trepel J, Aldape K, Pommier Y, Wu J. MGMT Status as a Clinical Biomarker in Glioblastoma. Trends Cancer 2020; 6:380-391. [PMID: 32348734 DOI: 10.1016/j.trecan.2020.02.010] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/07/2020] [Accepted: 02/13/2020] [Indexed: 12/27/2022]
Abstract
Glioblastoma is the most common primary malignant brain tumor. Although current standard therapy extends median survival to ~15 months, most patients do not have a sustained response to treatment. While O6-methylguanine (O6-MeG)-DNA methyltransferase (MGMT) promoter methylation status is accepted as a prognostic and promising predictive biomarker in glioblastoma, its value in informing treatment decisions for glioblastoma patients remains debatable. Discrepancies between MGMT promoter methylation status and treatment response in some patients may stem from inconsistencies between MGMT methylation and expression levels in glioblastoma. Here, we discuss MGMT as a biomarker and elucidate the discordance between MGMT methylation, expression, and patient outcome, which currently challenges the implementation of this biomarker in clinical practice.
Collapse
Affiliation(s)
- Madison Butler
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Lorinc Pongor
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yu-Ting Su
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Liqiang Xi
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Mark Raffeld
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Martha Quezado
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Jane Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yves Pommier
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | - Jing Wu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
10
|
Braczynski AK, Capper D, Jones DTW, Schittenhelm J, Stichel D, von Deimling A, Harter PN, Mittelbronn M. High density DNA methylation array is a reliable alternative for PCR-based analysis of the MGMT promoter methylation status in glioblastoma. Pathol Res Pract 2019; 216:152728. [PMID: 31784096 DOI: 10.1016/j.prp.2019.152728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/08/2019] [Accepted: 11/10/2019] [Indexed: 10/25/2022]
Abstract
AIM MGMT promoter methylation status is an important biomarker predicting survival and response to chemotherapy in patients suffering from glioblastoma. Since new diagnostic methods such as methylome-based classification of brain tumors are more and more frequently performed, we aimed at comparing the suitability of calculating the MGMT promoter methylation status in a quantitative manner from the methylome profiling as compared to the classic gold standard assessment by PCR. METHODS Our cohort consisted of 39 cases diagnosed as "glioblastoma, IDH-wildtype" of which the MGMT promoter methylation status was analyzed with both methylation-specific PCR and high density DNA methylation array using the STP-27 algorithm. Contradictory results were validated by pyrosequencing. RESULTS The inter-method reliability reached 77% (kappa-coefficient: 0.58) when also cases with an inconclusive result in one or the other method were taken into account. When only cases with conclusive results in both methods were considered, a very high inter-method reliability of 91% (kappa-coefficient: 0.86) could be achieved. For "methylated" cases, no contradictory results were obtained. For the remaining two cases with discrepant results subsequent pyrosequencing analyses spoke in favor of each previously applied method once. CONCLUSION In addition to its benefits for molecular subgrouping and copy number analysis of brain tumors, DNA-methylation based classification is a highly reliable tool for the assessment of MGMT promoter methylation status in glioblastoma patients.
Collapse
Affiliation(s)
- Anne K Braczynski
- Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany; Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany
| | - David Capper
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neuropathology, Charité Universitätsmedizin Berlin and German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - David T W Jones
- Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Jens Schittenhelm
- Department of Neuropathology, Institute of Pathology and Neuropathology, Eberhard-Karls University and Comprehensive Cancer Center Tuebingen-Stuttgart, Tuebingen, Germany
| | - Damian Stichel
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick N Harter
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
| | - Michel Mittelbronn
- Institute of Neurology (Edinger Institute), Goethe University, Frankfurt, Germany; NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health (LIH), Luxembourg; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg; National Center of Pathology (NCP), Laboratoire national de santé (LNS), Dudelange, Luxembourg; Luxembourg Centre of Neuropathology (LCNP), Luxembourg.
| |
Collapse
|
11
|
Storvall S, Ryhänen E, Heiskanen I, Vesterinen T, Bensch FV, Schildt J, Kytölä S, Karhu A, Arola J, Schalin-Jäntti C. MGMT Promoter Methylation and Parathyroid Carcinoma. J Endocr Soc 2019; 3:2114-2122. [PMID: 31687638 PMCID: PMC6821197 DOI: 10.1210/js.2019-00175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/17/2019] [Indexed: 11/19/2022] Open
Abstract
Context Parathyroid carcinoma (PC) is extremely rare. Prognosis is poor, with no known evidence-based systemic therapies. We previously reported complete remission in a patient with metastasized parathyroid carcinoma and high tumor MGMT promoter methylation status who was treated with temozolomide. Objective To study MGMT promoter methylation status in an additional set of aggressive parathyroid tumors. Design/Setting The study included 12 patients: 7 with sporadic and 5 with familial primary hyperparathyroidism (two of the latter carried a CDC73 gross deletion). Patient 9 is the previously described patient with PC and high MGMT methylation status. Her daughter (patient 12) had surgery for severe primary hyperparathyroidism due to atypical parathyroid adenoma during pregnancy. Eleven patients thus had PC and one had atypical parathyroid adenoma. MGMT promoter methylation status was determined from DNA extracted from primary (n = 10) or metastatic (n = 2) tumors. A mean methylation level >20% was considered high. Patient 11 had metastatic PC and received temozolomide cycles. Results Only the previously published patient (patient 9) had high tumor MGMT promoter methylation status. This was not a characteristic of the atypical parathyroid adenoma of the daughter (patient 12). Patient 11 (CDC73 intragenic deletion) has disseminated PC, low MGMT promoter methylation, and stable disease on follow-up after temozolomide treatment. Conclusion High MGMT promoter methylation status seems rare in PC. However, as demonstrated in other neuroendocrine tumors, some patients with disseminated PC might benefit from temozolomide. Demonstration of high methylation status could be a predictor of positive response to temozolomide treatment.
Collapse
Affiliation(s)
- Sara Storvall
- Department of Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eeva Ryhänen
- Department of Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ilkka Heiskanen
- Department of Surgery, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tiina Vesterinen
- Department of Pathology, HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Frank V Bensch
- Department of Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jukka Schildt
- HUS Medical Imaging Center, Department of Clinical Physiology and Nuclear Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Soili Kytölä
- Laboratory of Genetics, HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Auli Karhu
- Laboratory of Genetics, HUSLAB, Helsinki University Hospital, Helsinki, Finland.,Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.,Department of Applied Tumor Genomics, Research Programs Unit, University of Helsinki, Helsinki Finland
| | - Johanna Arola
- Department of Pathology, HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Camilla Schalin-Jäntti
- Department of Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
12
|
McAleenan A, Howell A, Kernohan A, Faulkner CL, Dawson S, Wragg C, Jefferies S, Brandner S, Vale L, Higgins JPT, Kurian KM. Prognostic value of test(s) for O 6
-methylguanine-DNA methyltransferase (MGMT) promoter methylation for predicting overall survival in people with glioblastoma treated with temozolomide. Hippokratia 2019. [DOI: 10.1002/14651858.cd013316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Alexandra McAleenan
- University of Bristol; Population Health Sciences, Bristol Medical School; 39 Whatley Road Bristol UK BS8 2PS
| | - Amy Howell
- University of Bristol; Health Sciences; Oakfield House Bristol UK BS8 2BN
| | - Ashleigh Kernohan
- Newcastle University; Institute of Health & Society; Baddiley-Clark Building, Richardson Road Newcastle upon Tyne UK NE2 4AA
| | - Claire L Faulkner
- Southmead Hospital; Bristol Genetics Laboratory, Pathology Sciences; North Bristol NHS Trust Bristol UK BS10 5NB
| | - Sarah Dawson
- University of Bristol; Population Health Sciences, Bristol Medical School; 39 Whatley Road Bristol UK BS8 2PS
| | - Christopher Wragg
- Southmead Hospital; Bristol Genetics Laboratory, Pathology Sciences; North Bristol NHS Trust Bristol UK BS10 5NB
| | - Sarah Jefferies
- Addenbrooke's Hospital; Department of Oncology; Hills Road Cambridge UK CB2 0QQ
| | - Sebastian Brandner
- The National Hospital for Neurology and Neurosurgery; Division of Neuropathology and Department of Neurodegeneration; University College Hospital NHS Foundation Trust and UCL Institute of Neurology Queen Square London UK WC1N 3BG
| | - Luke Vale
- Newcastle University; Institute of Health & Society; Baddiley-Clark Building, Richardson Road Newcastle upon Tyne UK NE2 4AA
| | - Julian P T Higgins
- University of Bristol; Population Health Sciences, Bristol Medical School; 39 Whatley Road Bristol UK BS8 2PS
| | - Kathreena M Kurian
- University of Bristol; Bristol Medical School: Brain Tumour Research Centre, Public Health Sciences; Oakfield House, Oakfield Grove Bristol UK BS8 2BN
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Duan S, Li M, Wang Z, Wang L, Liu Y. H19 induced by oxidative stress confers temozolomide resistance in human glioma cells via activating NF-κB signaling. Onco Targets Ther 2018; 11:6395-6404. [PMID: 30323617 PMCID: PMC6174297 DOI: 10.2147/ott.s173244] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Recent findings around long noncoding RNAs (lncRNAs) have opened novel areas of research around their prospective use in overcoming chemoresistance. Herein, we aimed to investigate the role of lncRNA H19 in temozolomide (TMZ) resistance of human glioma cells and the possible mechanisms. METHODS Short-/long-term oxidative stress was induced, and TMZ-resistant glioma cells (U251TMZ and LN229TMZ) were established. Small interfering RNA (siRNA) and overexpression plasmids were used to modulate the expression of H19 and/or luciferase the reporters. The MTT assay and immunoblotting of cleaved caspase-3, cyclin D1, XIAP and Bcl-2 were conducted to evaluate TMZ sensitivity. Luciferase reporter and quantitative real-time PCR (qRT-PCR) assays were used to verify the activation of NF-κB pathways by H19. RESULTS Knockdown of H19 in U251TMZ and LN229TMZ cells decreased half maximal inhibitory concentration (IC50) values for TMZ and increased cell apoptosis, and H19 overexpression in U251 and LN229 cells led to the opposite effects, indicating that the H19 confers TMZ resistance to glioma cells. Furthermore, knockdown of H19 decreased the NF-κB signaling, which was revealed by repressed reporter activity and declined expression of its downstream targets in TMZ-resistant glioma cells. In contrast, H19 overexpression in U251 and LN229 cells resulted in an increase in NF-κB activation. Blockage of NF-κB activation by its inhibitor abolished TMZ resistance caused by H19 overexpression. Addition of H2O2 to induce oxidative stress largely reversed TMZ sensitivity caused by H19 knockdown. CONCLUSION H19 confers TMZ resistance through activating NF-κB signaling and may represent a novel therapeutic target for TMZ-resistant gliomas.
Collapse
Affiliation(s)
- Shibo Duan
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, People's Republic of China,
| | - Ming Li
- Department of Obstetrics, Cangzhou Central Hospital, Cangzhou, Hebei Province, People's Republic of China
| | - Zhifeng Wang
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, People's Republic of China,
| | - Longlong Wang
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, People's Republic of China,
| | - Yongjie Liu
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, People's Republic of China,
| |
Collapse
|
16
|
Mao L, Whitehead CA, Paradiso L, Kaye AH, Morokoff AP, Luwor RB, Stylli SS. Enhancement of invadopodia activity in glioma cells by sublethal doses of irradiation and temozolomide. J Neurosurg 2018; 129:598-610. [DOI: 10.3171/2017.5.jns17845] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVEGlioblastoma is the most common primary central nervous system tumor in adults. These tumors are highly invasive and infiltrative and result in tumor recurrence as well as an extremely poor patient prognosis. The current standard of care involves surgery, radiotherapy, and chemotherapy. However, previous studies have suggested that glioblastoma cells that survive treatment are potentially more invasive. The goal of this study was to investigate whether this increased phenotype in surviving cells is facilitated by actin-rich, membrane-based structures known as invadopodia.METHODSA number of commercially available cell lines and glioblastoma cell lines obtained from patients were initially screened for the protein expression levels of invadopodia regulators. Gelatin-based zymography was also used to establish their secretory protease profile. The effects of radiation and temozolomide treatment on the glioblastoma cells were then investigated with cell viability, Western blotting, gelatin-based zymography, and invadopodia matrix degradation assays.RESULTSThe authors’ results show that the glioma cells used in this study express a number of invadopodia regulators, secrete MMP-2, and form functional matrix-degrading invadopodia. Cells that were treated with radiotherapy and temozolomide were observed to show an increase primarily in the activation of MMP-2. Importantly, this also resulted in a significant enhancement in the invadopodia-facilitated matrix-degrading ability of the cells, along with an increase in the percentage of cells with invadopodia after radiation and temozolomide treatment.CONCLUSIONSThe data from this study suggest that the increased invasive phenotype that has been previously observed in glioma cells posttreatment is mediated by invadopodia. The authors propose that if the formation or activity of these structures can be disrupted, they could potentially serve as a viable target for developing novel adjuvant therapeutic strategies that can be used in conjunction with the current treatment protocols in combatting the invasive phenotype of this deadly disease.
Collapse
Affiliation(s)
- Leon Mao
- 1Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital; and
| | - Clarissa A. Whitehead
- 1Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital; and
| | - Lucia Paradiso
- 1Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital; and
| | - Andrew H. Kaye
- 1Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital; and
- 2Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Andrew P. Morokoff
- 1Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital; and
- 2Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Rodney B. Luwor
- 1Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital; and
| | - Stanley S. Stylli
- 1Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital; and
- 2Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
17
|
Akhtar S, Vranic S, Cyprian FS, Al Moustafa AE. Epstein-Barr Virus in Gliomas: Cause, Association, or Artifact? Front Oncol 2018; 8:123. [PMID: 29732319 PMCID: PMC5919939 DOI: 10.3389/fonc.2018.00123] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/04/2018] [Indexed: 12/17/2022] Open
Abstract
Gliomas are the most common malignant brain tumors and account for around 60% of all primary central nervous system cancers. Glioblastoma multiforme (GBM) is a grade IV glioma associated with a poor outcome despite recent advances in chemotherapy. The etiology of gliomas is unknown, but neurotropic viruses including the Epstein–Barr virus (EBV) that is transmitted via salivary and genital fluids have been implicated recently. EBV is a member of the gamma herpes simplex family of DNA viruses that is known to cause infectious mononucleosis (glandular fever) and is strongly linked with the oncogenesis of several cancers, including B-cell lymphomas, nasopharyngeal, and gastric carcinomas. The fact that EBV is thought to be the causative agent for primary central nervous system (CNS) lymphomas in immune-deficient patients has led to its investigations in other brain tumors including gliomas. Here, we provide a review of the clinical literature pertaining to EBV in gliomas and discuss the possibilities of this virus being simply associative, causative, or even an experimental artifact. We searched the PubMed/MEDLINE databases using the following key words such as: glioma(s), glioblastoma multiforme, brain tumors/cancers, EBV, and neurotropic viruses. Our literature analysis indicates conflicting results on the presence and role of EBV in gliomas. Further comprehensive studies are needed to fully implicate EBV in gliomagenesis and oncomodulation. Understanding the role of EBV and other oncoviruses in the etiology of gliomas, would likely open up new avenues for the treatment and management of these, often fatal, CNS tumors.
Collapse
Affiliation(s)
| | - Semir Vranic
- College of Medicine, Qatar University, Doha, Qatar
| | | | - Ala-Eddin Al Moustafa
- College of Medicine, Qatar University, Doha, Qatar.,Biomedical Research Centre, Qatar University, Doha, Qatar.,Oncology Department, McGill University, Montreal, QC, Canada
| |
Collapse
|
18
|
Jiménez D, Matamala JM, Chiti A, Vergara C, Tissera C, Melo R, Cartier L. O 6-methylguanine-DNA-methyltransferase immunostaining intensity in glioblastoma. Neurol Neurochir Pol 2017; 52:116-119. [PMID: 29153917 DOI: 10.1016/j.pjnns.2017.10.014] [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: 08/18/2017] [Revised: 10/10/2017] [Accepted: 10/25/2017] [Indexed: 11/16/2022]
Abstract
Immunohistochemistry (IHC) for O6-methylguanine-DNA-methyltransferase (MGMT) has shown heterogeneous results. Cell staining intensity has not been included as a quantifiable variable in IHC analyses. We performed MGMT IHC in 29 patients diagnosed as glioblastoma classifying cells into three categories based on nuclear staining intensity compared with adjacent endothelium. The median proportions of strong-moderate, weak and no staining cells were 10%, 16% and 71%, respectively. The proportion of positive cases for MGMT expression varies from 38% to 52% depending on the classification of weakly stained cells. This letter challenges previous studies that have not included intensity as a variable for IHC analysis.
Collapse
Affiliation(s)
- Daniel Jiménez
- Laboratory of Biomedical Sciences, Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago, Chile; Neurology Service, Hospital del Salvador, Santiago, Chile.
| | - José Manuel Matamala
- Laboratory of Biomedical Sciences, Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago, Chile; Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alessandra Chiti
- Laboratory of Biomedical Sciences, Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Carmen Vergara
- Laboratory of Biomedical Sciences, Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | - Romulo Melo
- Instituto de Neurocirugía Dr. Asenjo, Santiago, Chile; Department of Neurological Science, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luis Cartier
- Laboratory of Biomedical Sciences, Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago, Chile; Neurology Service, Hospital del Salvador, Santiago, Chile
| |
Collapse
|
19
|
Shi J, Dong B, Zhou P, Guan W, Peng Y. Functional network analysis of gene-phenotype connectivity associated with temozolomide. Oncotarget 2017; 8:87554-87567. [PMID: 29152101 PMCID: PMC5675653 DOI: 10.18632/oncotarget.20848] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/17/2017] [Indexed: 02/07/2023] Open
Abstract
Rationale Glioma has a poor survival rate in patients even with aggressive treatment. Temozolomide (TMZ) is the standard chemotherapeutic choice for treating glioma, but TMZ treatment consistently leads to high resistance. Aim To investigate the underlying mechanisms of TMZ action with new therapeutic regimens in glioma. Methods and results The biological effects of TMZ mainly depend on the three following DNA repair systems: methylguanine methyltransferase (MGMT), mismatch repair (MMR) and base excision repair (BER). Based on related genes in these three systems, web-based tools containing data compiled from open-source databases, including DrugBank, STRING, WebGestalt and ClueGO, were queried, and five common genes along with the top fifteen pathways, including the glioma pathway, were identified. A genomic analysis of the six genes identified in the glioma pathway by cBioPortal indicated that TMZ might exert biological effects via interaction with the tumor protein P53(TP53) signaling axis. Finally, a survival analysis with the six genes in glioma cases (low-grade glioma and glioblastoma multiforme) was conducted using OncoLnc, which might provide directions for the future exploration of prognosis in glioma. Conclusions This study indicates that a functional network analysis resembles a "BioGPS", with the ability to draw a web-based scientific map that can productively and cost-effectively associate TMZ with its primary and secondary biological targets.
Collapse
Affiliation(s)
- Jia Shi
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| | - Bo Dong
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| | - Peng Zhou
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| | - Wei Guan
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| | - Ya Peng
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou City, 213003, China
| |
Collapse
|
20
|
Wu J, Zhang H, Xu Y, Zhang J, Zhu W, Zhang Y, Chen L, Hua W, Mao Y. Juglone induces apoptosis of tumor stem-like cells through ROS-p38 pathway in glioblastoma. BMC Neurol 2017; 17:70. [PMID: 28388894 PMCID: PMC5383964 DOI: 10.1186/s12883-017-0843-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 03/20/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Juglone is a natural pigment, which has cytotoxic effect against various human tumor cells. However, its cytotoxicity to glioma cells, especially to tumor stem-like cells (TSCs) has not been demonstrated. METHODS TSCs of glioma were enriched from U87 and two primary cells (SHG62, and SHG66) using serum-free medium supplemented with growth factors, including bFGF, EGF and B27. After treatment of juglone with gradient concentrations (0, 10, 20, and 40 μM), the viability and apoptosis of TSCs were evaluated by WST-8 assay and flow cytometry. Reactive oxygen species (ROS) was labeled by the cell-permeable fluorescent probe and detected with flow cytometry. ROS scavenger (NAC) and p38-MAPK inhibitor (SB203580) were applied to resist the cytotoxic effect. Caspase 9 cleavage and p38 phosphorylation (P-p38) were quantified by western blot. Juglone as well as temozolomide (TMZ) were administrated in intracranial xenografts and MR scan was performed every week to evaluate the anti-tumor effect in vivo. RESULTS Juglone could obviously inhibit the proliferation of TSCs in glioma by decreasing cell viability (P < 0.01) and inducing apoptosis (P < 0.01), which was accompanied by increased caspase 9 cleavage in a dose-dependent manner (P < 0.01). In the meantime, juglone could generate ROS significantly and increase p38 phosphorylation (P < 0.01). In addition, pretreatment with ROS scavenger or p38-MAPK inhibitor could reverse juglone-induced cytotoxicity (P < 0.01). More importantly, juglone could also suppress tumor growth in vivo and improve the survival of U87-bearing mice compared with control (P < 0.05), although TMZ seemed to have better effect. CONCLUSIONS Juglone could inhibit the growth of TSCs in gliomas through the activation of ROS-p38-MAPK pathway in vitro, and the anti-glioma effect was validated in vivo, which offers a potential therapeutic agent to gliomas.
Collapse
Affiliation(s)
- Jinfeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, #12 Middle Wurumuqi Road, Shanghai, 200040, People's Republic of China
| | - Haibo Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, #12 Middle Wurumuqi Road, Shanghai, 200040, People's Republic of China
| | - Yang Xu
- Department of Neurosurgery, Huashan Hospital, Fudan University, #12 Middle Wurumuqi Road, Shanghai, 200040, People's Republic of China
| | - Jingwen Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, #12 Middle Wurumuqi Road, Shanghai, 200040, People's Republic of China.,Department of Ultrasound, Hebei General Hospital, #348 West Heping Road, Shijiazhuang, Hebei Province, 050000, People's Republic of China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, #12 Middle Wurumuqi Road, Shanghai, 200040, People's Republic of China
| | - Yi Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, #12 Middle Wurumuqi Road, Shanghai, 200040, People's Republic of China
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, #12 Middle Wurumuqi Road, Shanghai, 200040, People's Republic of China.
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, #12 Middle Wurumuqi Road, Shanghai, 200040, People's Republic of China.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, #12 Middle Wurumuqi Road, Shanghai, 200040, People's Republic of China.,Institutes of Biomedical Sciences, Fudan University, #131 Dong'an Road, Shanghai, 200040, People's Republic of China.,State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, 200040, People's Republic of China.,The Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200040, People's Republic of China
| |
Collapse
|
21
|
Gzell C, Back M, Wheeler H, Bailey D, Foote M. Radiotherapy in Glioblastoma: the Past, the Present and the Future. Clin Oncol (R Coll Radiol) 2017; 29:15-25. [DOI: 10.1016/j.clon.2016.09.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/22/2016] [Accepted: 08/25/2016] [Indexed: 10/25/2022]
|
22
|
Creemers SG, van Koetsveld PM, van den Dungen ESR, Korpershoek E, van Kemenade FJ, Franssen GJH, de Herder WW, Feelders RA, Hofland LJ. Inhibition of Human Adrenocortical Cancer Cell Growth by Temozolomide in Vitro and the Role of the MGMT Gene. J Clin Endocrinol Metab 2016; 101:4574-4584. [PMID: 27603910 PMCID: PMC5155680 DOI: 10.1210/jc.2016-2768] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Treatment of patients with adrenocortical carcinomas (ACC) with mitotane and/or chemotherapy is often associated with toxicity and poor tumor response. New therapeutic options are urgently needed. OBJECTIVE The objectives of the study were to evaluate the therapeutic possibilities of temozolomide (TMZ) in ACC cells and to assess the potential predictive role of the DNA repair gene O6-Methylguanine-DNA methyltransferase (MGMT) in adrenocortical tumors. METHODS Three human ACC cell lines and eight primary ACC cultures were used to assess effects of TMZ in vitro. In the cell lines, 11 normal adrenals, 16 adrenocortical adenomas, and 29 ACC, MGMT promoter methylation and expression were determined. RESULTS IC50 values of TMZ on cell growth were 39 μM, 38 μM, and 44 μM for H295R, HAC15, and SW13, respectively. TMZ induced apoptosis and provoked cytotoxic and cytostatic effects by reducing the surviving fraction of ACC colonies and the colony size. TMZ thereby induced cell cycle arrests in ACC cell lines. TMZ and mitotane both inhibited growth of ACC cells cultured as three-dimensional spheroids. TMZ inhibited cell amount in five of eight primary ACC cultures and induced apoptosis in seven of eight primary ACC cultures. In ACC cell lines and adrenal tissues, MGMT promoter methylation was low. In ACCs, methylation was inversely correlated with MGMT mRNA expression. MGMT protein expression was not correlated with MGMT methylation. CONCLUSIONS For the first time, we show the therapeutic potential of temozolomide for ACC, offering an urgently needed potential alternative for patients not responding to mitotane alone or with etoposide, doxorubicin, and cisplatin. (Pre-)clinical studies are warranted to assess efficacy in vivo.
Collapse
Affiliation(s)
- S G Creemers
- Department of Internal Medicine, Division of Endocrinology (S.G.C., P.M.v.K., E.S.R.v.d.D., W.W.d.H., R.A.F., L.J.H.), and Departments of Pathology (E.K., F.J.v.K.) and Surgery (G.J.H.F.), Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands
| | - P M van Koetsveld
- Department of Internal Medicine, Division of Endocrinology (S.G.C., P.M.v.K., E.S.R.v.d.D., W.W.d.H., R.A.F., L.J.H.), and Departments of Pathology (E.K., F.J.v.K.) and Surgery (G.J.H.F.), Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands
| | - E S R van den Dungen
- Department of Internal Medicine, Division of Endocrinology (S.G.C., P.M.v.K., E.S.R.v.d.D., W.W.d.H., R.A.F., L.J.H.), and Departments of Pathology (E.K., F.J.v.K.) and Surgery (G.J.H.F.), Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands
| | - E Korpershoek
- Department of Internal Medicine, Division of Endocrinology (S.G.C., P.M.v.K., E.S.R.v.d.D., W.W.d.H., R.A.F., L.J.H.), and Departments of Pathology (E.K., F.J.v.K.) and Surgery (G.J.H.F.), Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands
| | - F J van Kemenade
- Department of Internal Medicine, Division of Endocrinology (S.G.C., P.M.v.K., E.S.R.v.d.D., W.W.d.H., R.A.F., L.J.H.), and Departments of Pathology (E.K., F.J.v.K.) and Surgery (G.J.H.F.), Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands
| | - G J H Franssen
- Department of Internal Medicine, Division of Endocrinology (S.G.C., P.M.v.K., E.S.R.v.d.D., W.W.d.H., R.A.F., L.J.H.), and Departments of Pathology (E.K., F.J.v.K.) and Surgery (G.J.H.F.), Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands
| | - W W de Herder
- Department of Internal Medicine, Division of Endocrinology (S.G.C., P.M.v.K., E.S.R.v.d.D., W.W.d.H., R.A.F., L.J.H.), and Departments of Pathology (E.K., F.J.v.K.) and Surgery (G.J.H.F.), Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands
| | - R A Feelders
- Department of Internal Medicine, Division of Endocrinology (S.G.C., P.M.v.K., E.S.R.v.d.D., W.W.d.H., R.A.F., L.J.H.), and Departments of Pathology (E.K., F.J.v.K.) and Surgery (G.J.H.F.), Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands
| | - L J Hofland
- Department of Internal Medicine, Division of Endocrinology (S.G.C., P.M.v.K., E.S.R.v.d.D., W.W.d.H., R.A.F., L.J.H.), and Departments of Pathology (E.K., F.J.v.K.) and Surgery (G.J.H.F.), Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands
| |
Collapse
|
23
|
Yaghmour G, Prouet P, Wiedower E, Jamy OH, Feldman R, Chandler JC, Pandey M, Martin MG. Genomic alterations in neuroendocrine cancers of the ovary. J Ovarian Res 2016; 9:52. [PMID: 27566252 PMCID: PMC5002197 DOI: 10.1186/s13048-016-0259-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/11/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND As we have previously reported, small cell carcinoma of the ovary (SCCO) is a rare, aggressive form of ovarian cancer associated with poor outcomes. In an effort to identify new treatment options, we utilized comprehensive genomic profiling to assess the potential for novel therapies in SCCO. METHODS Patients with SCCO, SCCO-HT (hypercalcemic type), neuroendocrine tumors of the ovary (NET-O), and small cell carcinoma of the lung (SCLC) profiled by Caris Life Sciences between 2007-2015 were identified. Tumors were assessed with up to 21 IHC stains, in situ hybridization of cMET, EGFR, HER2 and PIK3CA, and next-generation sequencing (NGS) as well as Sanger sequencing of selected genes. RESULTS Forty-six patients with SCCO (10 SCCO, 18 SCCO-HT, 18 NET-O) were identified as well as 58 patients with SCLC for comparison. Patients with SCCO and SCCO-HT were younger (median 42 years [range 12-75] and 26 years [range 8-40], respectively) than patients with NET-O 62 [range 13-76] or SCLC 66 [range 36-86]. SCCO patients were more likely to be metastatic (70 %) than SCCO-HT (50 %) or NET-O (33 %) patients, but at a similar rate to SCLC patients (65 %). PD1 expression varied across tumor type with SCCO (100 %), SCCO-HT (60 %), NET-O (33 %) vs SCLC (42 %). PDL1 expression also varied with SCCO (50 %), SCCO-HT (20 %), NET-O (33 %) and SCLC (0 %). No amplifications were identified in cMET, EGFR, or HER2 and only 1 was found in PIK3CA (NET-O). Actionable mutations were rare with 1 patient with SCCO having a BRCA2 mutation and 1 patient with NET-O having a PIK3CA mutation. No other actionable mutations were identified. CONCLUSIONS No recurrent actionable mutations or rearrangements were identified using this platform in SCCO. IHC patterns may help guide the use of chemotherapy in these rare tumors.
Collapse
Affiliation(s)
- George Yaghmour
- The West Cancer Center, 1588 Union Ave., Memphis, TN 38104 USA
- Department of Hematology & Oncology, The University of Tennessee Health Science Center, 956 Court Ave., Suite H310A, Memphis, TN 38163 USA
| | - Philippe Prouet
- Department of Internal Medicine, The University of Tennessee Health Science Center, 956 Court Ave., Suite H314, Memphis, TN 38163 USA
| | - Eric Wiedower
- The West Cancer Center, 1588 Union Ave., Memphis, TN 38104 USA
- Department of Hematology & Oncology, The University of Tennessee Health Science Center, 956 Court Ave., Suite H310A, Memphis, TN 38163 USA
| | - Omer Hassan Jamy
- Department of Internal Medicine, The University of Tennessee Health Science Center, 956 Court Ave., Suite H314, Memphis, TN 38163 USA
| | - Rebecca Feldman
- Caris Life Sciences, 4750 S. 44th Place, Phoenix, AZ 85040 USA
| | - Jason C Chandler
- The West Cancer Center, 1588 Union Ave., Memphis, TN 38104 USA
- Department of Hematology & Oncology, The University of Tennessee Health Science Center, 956 Court Ave., Suite H310A, Memphis, TN 38163 USA
| | - Manjari Pandey
- The West Cancer Center, 1588 Union Ave., Memphis, TN 38104 USA
- Department of Hematology & Oncology, The University of Tennessee Health Science Center, 956 Court Ave., Suite H310A, Memphis, TN 38163 USA
| | - Mike G Martin
- The West Cancer Center, 1588 Union Ave., Memphis, TN 38104 USA
- Department of Hematology & Oncology, The University of Tennessee Health Science Center, 956 Court Ave., Suite H310A, Memphis, TN 38163 USA
| |
Collapse
|
24
|
Smith AW, Parashar B, Wernicke AG. Subventricular zone-associated glioblastoma: A call for translational research to guide clinical decision making. NEUROGENESIS 2016; 3:e1225548. [PMID: 27900341 DOI: 10.1080/23262133.2016.1225548] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 07/21/2016] [Accepted: 08/14/2016] [Indexed: 10/21/2022]
Abstract
Glioblastoma (GBM) is both the most common and the most devastating primary cancer of the central nervous system, with an expected overall survival in most patients of about 14 months. Despite extensive research, outcomes for GBM have been largely unchanged since the introduction of temozolomide in 2005. We believe that in order to achieve a breakthrough in therapeutic management, we must begin to identify subtypes of GBM, and tailor treatment to best target a particular tumor's vulnerabilities. Our group has recently produced an examination of the clinical outcomes of radiation therapy directed at tumors that contact the subventricular zone (SVZ), the 3-5 mm lateral border of the lateral ventricles that contains the largest collection of neural stem cells in the adult brain. We find that SVZ-associated tumors have worse progression free and overall survival than tumors that do not contact the SVZ, and that they exhibit unique recurrence and migration patterns. However, with minimal basic science research into SVZ-associated GBM, it is currently impossible to determine if the clinicobehavioral uniqueness of this group of tumors represents a true disease subtype from a genetic perspective. We believe that further translational research into SVZ-associated GBM is needed to establish a therapeutic profile.
Collapse
Affiliation(s)
- Andrew W Smith
- University of Rochester School of Medicine and Dentistry , Rochester, NY, USA
| | - Bhupesh Parashar
- Stitch Radiation Oncology, Weill-Cornell Medical College/New York Presbyterian Hospital , New York, NY, USA
| | - A Gabriella Wernicke
- Stitch Radiation Oncology, Weill-Cornell Medical College/New York Presbyterian Hospital , New York, NY, USA
| |
Collapse
|