Stereotactic image‑based histological analysis reveals a correlation between 11C‑methionine uptake and MGMT promoter methylation in non‑enhancing gliomas

  • Authors:
    • Yoshiko Okita
    • Tomoko Shofuda
    • Daisuke Kanematsu
    • Ema Yoshioka
    • Yoshinori Kodama
    • Masayuki Mano
    • Manabu Kinoshita
    • Masahiro Nonaka
    • Shin Nakajima
    • Toshiyuki Fujinaka
    • Yonehiro Kanemura
  • View Affiliations

  • Published online on: May 31, 2018     https://doi.org/10.3892/ol.2018.8866
  • Pages: 1924-1930
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gliomas are genetically and histopathologically heterogeneous. Intratumoral heterogeneity in the MGMT promoter methylation status is an important clinical biomarker of glioblastoma. A higher uptake of 11C‑methionine in positron‑emission tomography (PET) reportedly reflects increased MGMT promoter methylation; however, non‑stereotactic comparison of MGMT methylation and 11C‑methionine PET images may not be accurate. The present study examined the correlation between 11C‑methionine uptake and MGMT promoter methylation in non‑enhancing gliomas using stereotactic image‑based histological analysis. Data were collected from 9 patients with newly diagnosed non‑enhancing glioma who underwent magnetic resonance imaging and 11C‑methionine PET during pre‑surgical examination. Clinical data were also collected from 3 patients during repeat surgery. The correlation between 11C‑methionine uptake and MGMT methylation or cell density was analyzed using histological specimens obtained by multiple stereotactic sampling and an exact local comparison of 11C‑methionine PET images and histological specimens was made. A total of 31 stereotactic sample sites were identified. In newly diagnosed cases, the tumor to normal uptake (T/N) ratio revealed a significant positive correlation with MGMT methylation (R=0.54, P=0.009) and a marginal correlation with cell density (R=0.42, P=0.05). In recurrent cases, the T/N ratio demonstrated no correlation with MGMT methylation (R=0.01, P=0.97) or cell density (R=0.15, P=0.70). An increased uptake of 11C‑methionine in PET may reflect increased MGMT promoter methylation according to stereotactic image‑based histological analysis. 11C‑methionine PET could therefore be a useful tool for detecting regional MGMT promoter methylation in non‑enhancing primary glioma.

Introduction

MGMT promoter methylation is associated with a favorable outcome after temozolomide chemotherapy in patients with newly diagnosed glioblastoma (1). Furthermore, temozolomide rechallenge is a treatment option for recurrent MGMT promoter-methylated glioblastoma (2). Evaluation of MGMT methylation status is thus important for treatment of primary and recurrent glioma.

Gliomas are genetically and histopathologically heterogeneous (35). A previous study has demonstrated intratumoral heterogeneity in MGMT promoter methylation status (6). Therefore, it is doubtful that a single biopsy specimen can represent the molecular landscape of the entire tumor.

MGMT promoter methylation status can change between the first surgery for newly diagnosed glioblastoma and a second surgery for recurrent disease (7,8). The development of reduced methylation in the MGMT promoter leads to acquired therapeutic resistance after temozolomide treatment in glioblastomas (9). Re-evaluation of MGMT promoter methylation status at the time of recurrence is therefore important for selecting treatment.

Attempts to assess gene mutations non-invasively by imaging technology have been conducted in the past, but there are few reports on image evaluation of glioma-related gene mutations (1015). However, among these, 11C-methionine positron-emission tomography (11C-methionine PET) has been proven to be a useful tool for detecting MGMT promoter methylation in non-enhancing glioma (12). Nevertheless, given the heterogeneity of MGMT promoter methylation status in gliomas, a non-stereotactic comparison of MGMT promoter methylation and 11C-methionine uptake in PET images may not be accurate.

In this study, the correlation between 11C-methionine PET and MGMT promoter methylation status in non-enhancing gliomas was stereotactically verified.

Materials and methods

Subjects and glioma tumor samples

The present study was performed in accordance with the principles of the Helsinki Declaration, and approval for this study was obtained from the ethical committee of Osaka National Hospital (no. 94, IRB no. 0713). All patients provided written informed consent.

Clinical data were collected from 9 patients with newly diagnosed glioma, who underwent both magnetic resonance imaging (MRI) and 11C-methionine PET as part of their pre-surgical examination, from 2014 to 2017. Clinical data were also collected from 3 of 9 patients with newly diagnosed glioma during surgery for recurrent disease. Recurrent cases received chemotherapy or chemoradiotherapy prior to 11C-methionine PET examination. Tumor tissue specimens were also obtained from each patient. Multiple tissue sites were stereotactically sampled, and each sample was evenly divided. One part of each sample was subjected to pathological examination and the other was subjected to MGMT methylation status analysis.

Histopathological analysis

A subset of the tumor samples was fixed in 10% formalin and embedded in paraffin wax following standard procedures. In each case, hematoxylin and eosin (H&E)-stained sections were examined to classify the tumors according to the World Health Organization (WHO) International Histological Classification of Tumors. Cell counting was performed under a light microscope (Olympus Corporation, Tokyo, Japan; ×200 magnification) and data were recorded as the mean of 3 different locations within the specimen according to the method used in our previous study (16).

Genomic DNA Extraction

Tumor samples were immediately frozen in liquid nitrogen and stored at −80°C. Genomic DNA was extracted using the DNeasy Blood and Tissue kit (Qiagen, Inc., Valencia, CA, USA), according to the manufacturer's protocol (12).

MGMT Promoter Methylation Analysis

MGMT promoter methylation status was determined by quantitative methylation-specific PCR (qMSP) as described in our previous study (12). DNA extracted from tumor tissue was subjected to bisulfite modification by an EZ DNA Methylation-Gold kit (Zymo Research Corp., Irvine, CA, USA), according to the manufacturer's instructions. Bisulfite-modified DNA was analyzed by qMSP using the QuantStudio™ 12K Flex Real-Time PCR System (Thermo Fisher Scientific, Inc., Waltham, MA, USA) with POWER SYBR® Green PCR Master Mix (Thermo Fisher Scientific, Inc.). Methylated and unmethylated DNA molecules were amplified separately using specific primers (17). Quantification of methylated and unmethylated sequences was performed by employing the standard curve method, using serial dilutions of bisulfite-modified EpiScope® Methylated HCT116 gDNA (Takara Bio, Inc., Shiga, Japan), which is highly methylated by CpG methylase, and EpiScope® Unmethylated HCT116 DKO gDNA (Takara Bio, Inc.), obtained from cells that genetically lack both DNA methyltransferase 1 and DNA methyltransferase 3B. The percentages of methylation and standard deviations (S.D.) were calculated from triplicate PCRs.

11C-methionine PET

PET images were obtained using a SET-3000 GCT/X scanner (Shimadzu Corp., Kyoto, Japan) with gadolinium oxyorthosilicate crystals as emission detectors. The 11C-methionine tracer was synthesized in accordance with the method described by Berger et al (18) and injected intravenously at a dose of 111-222 mBq (3–6 mCi). Tracer accumulation was recorded over 15 min in 99 transaxial slices, spanning the entire brain. The summed activity at 20-35 min after tracer injection was used for image reconstruction. Images were stored in 256×256×99 anisotropic voxels, with a voxel size of 1×1×2.6 mm. The tumor/normal tissue (T/N) ratios were calculated stereotactically by dividing the standard uptake value (SUV) for the tumor by the SUV of the contralateral lesion in the same way as in our previous study (16).

Magnetic resonance imaging

All patients were studied using a 1.5 T whole-body MR system (Achieva; Philips Healthcare, Amsterdam, The Netherlands) within a week before the operation. T1-weighted imaging with gadolinium enhancement was used to select patients with non-enhancing gliomas. T2-weighted (T2) or FLAIR images were acquired in all cases for delineation of tumors.

Image fusion and registration

We used the Brainlab VectorVision compact neuronavigation system (Brainlab, Munich, Germany). This neuronavigation system is composed of a surgical planning workstation with software tools for coregistration of multimodal image sets. The PET image was registered on fluid-attenuated inversion recovery (FLAIR) standard anatomical MRI images using the Brainlab VectorVision compact neuronavigation system. These registered images were transferred to the navigation system for stereotactic surgery.

Surgery for stereotactic multiple sampling evaluation

We used a stereotactic multiple sampling evaluation for gliomas as previously described (16). The location for tumor biopsy was preoperatively determined on FLAIR images and 11C-methionine PET. 3D gapless FLAIR images and PET data, co-registered beforehand, were transferred to the Brainlab VectorVision compact neuronavigation system and the biopsy target for histopathological examination was planned (Fig. 1). Standard craniotomy was performed under general anesthesia. Multiple sampling biopsy was performed in non-enhancing tumor lesions targeted for resection by inserting a catheter, aimed at the target, immediately after craniotomy, in order to minimize the error caused by brain shifting. Although multiple tissue sampling was performed in some cases, real-time navigation was performed to confirm the position of each biopsy site within the tumor.

Statistical analysis

All data are presented as the mean value unless otherwise stated. Statistical analysis was performed using JMP version 8 (SAS Institute, Inc., Cary, NC, USA). A linear regression model, using the method for least squares, was used for modeling 2 or 3 independent variables. A P-value of <0.05 was considered statistically significant.

Results

Patient characteristics

Detailed information on the 9 investigated patients is listed in Table I. Tissue specimens at 31 sampling sites were stereotactically obtained using an intraoperative neuronavigation system. All patients had non-enhancing lesions on MRI.

Table I.

Caracteristics of patients with non-enhancing glioma.

Table I.

Caracteristics of patients with non-enhancing glioma.

Patient no.Age (years)SexHistologic diagnosisPrimary or recurrentTreatment before operationLocalizationNo. of specimensT/N ratioMGMT methylation (%)MGMT methylation status (M: ≥3%, U:<3%)
1–157FDiffuse astrocytomaPrimaryNoneR frontal11.1948.48M
21.3346.51M
31.3217.5M
1–259FGlioblastomaFirst recurrentTMZL frontal11.112.77U
21.4512.91M
2–179MDiffuse astrocytomaPrimaryNoneR parietal11.240.06U
21.060.03U
32.230.17U
2–281MAnaplastic astrocytomaFirst recurrentTMZR temporal10.960.38U
21.490.24U
2–381MAnaplastic astrocytomaSecond recurrentRT + TMZL parietal11.820.24U
21.520.2U
366M OligodendrogliomaPrimaryNoneL frontal11.0119.77M
21.348.16M
443M OligodendrogliomaPrimaryNoneL frontal11.382.56U
21.222.29U
31.292.61U
5–146F OligodendrogliomaPrimaryNoneR frontal11.9647.93M
22.4584.67M
5–242FAnaplastic oligodendrogliomaFirst recurrentTMZR frontal11.080.32U
20.591.34U
32.030.62U
631FAnaplastic astrocytomaPrimaryNoneR frontal11.031.6U
21.394.75M
30.951.39U
738MAnaplastic astrocytomaPrimaryNoneL frontal11.560.14U
21.420.21U
30.730.23U
858FGlioblastomaPrimaryNoneL temporal11.250.21U
950MGlioblastomaPrimaryNoneR occipital11.170.2U
21.160.33U

[i] M, male; F, female; R, right; L, left; RT, radiotherapy; TMZ, temozolomide; M, methylated; U, unmethylated; MGMT, O(6)-methylguanine-DNA methyltransferase; T/N, tumor to normal uptake ratio.

Intratumoral heterogeneity in MGMT promoter methylation status

In our previous report, we evaluated the correlation between T/N ratio and MGMT methylation in a non-stereotactic manner, and found that a threshold T/N ratio value of 1.6 significantly correlated with a quantitative threshold MGMT methylation status of 3% (12). Using a positive methylation assay threshold of 3, 22.2% (2 of 9) of cases in this study demonstrated intratumoral heterogeneity in terms of methylation levels, with the percentage of methylation varying up to 4.66-fold for each case (Table I).

Correlation of PET T/N 11C-methionine ratio with MGMT methylation and cell density in stereotactic image-based histological comparisons. In newly diagnosed cases, the T/N ratio correlated positively with MGMT methylation (R=0.54, P=0.009) (Fig. 2A). In contrast, the T/N ratio showed a marginal correlation with cell density (R=0.42, P=0.05) (Fig. 2B).

In recurrent cases, the T/N ratio did not correlate with MGMT methylation (R=0.01, P=0.97) or with cell density (R=0.15, P=0.70) (Fig. 3A and B).

Estimation of 11C-methionine PET with MGMT methylation and cell density

We attempted to model the PET T/N ratio of 11C-methionine according to MGMT methylation and cell density in newly diagnosed cases. A linear regression model indicated the following equation, with an overall P-value of 0.025, which was considered statistically significant for fit.

(T/Nr of 11C-methionie PET)=0.0002×CD+0.004×MGMT+1.1,

where CD is the cell density (cells/mm2) and MGMT indicates MGMT methylation (%). Multiple regression analysis revealed that MGMT methylation tended to be statistically significant for model construction (t=2.03, P=0.057), but cell density was not (t=0.87, P=0.4).

Discussion

Previously, we evaluated the highest T/N ratio of 11C-methionine PET in non-enhancing gliomas, in a non-stereotactic manner, and identified that a higher uptake of 11C-methionine in PET may reflect increased MGMT promoter methylation (12). However, gliomas are genetically and histopathologically heterogeneous (35). A previous report had demonstrated intratumoral heterogeneity in MGMT promoter methylation status in 14% of cases, with the percentage of methylation varying up to 4-fold within each case, based on pyrosequencing results (6). Thus, caution must be taken when interpreting the results of genomic biomarker analyses based on a single biopsy specimen from a given tumor. Therefore, in this study, we used stereotactic image-based histological analysis and identified the correlation between the T/N ratio and MGMT promoter methylation in multiple, spatially distinct samples of primary gliomas. Using qMSP, we found that 22.2% of cases demonstrated intratumoral heterogeneity in methylation levels.

11C-methionine uptake is mainly determined by a sodium-independent L-transporter system in the luminal membrane of endothelial cells (1921) and correlates with the proliferation and microvessel density of tumors (16,18,22,23). Although the main metabolic pathway of 11C-methionine is protein incorporation, the conversion of methionine to S-adenosyl-L-methionine (SAM) is a minor pathway (24). In the brain, methionine is a precursor of SAM (25) which plays an important role in DNA methylation processes (26). The activation of methionine to SAM apparently occurs very rapidly in the brain, as measured by the conversion of administered 11C-methionine, and 11C-SAM uptake may reflect the enhanced transmethylation processes in tumors (24,27). Methionine uptake in tumors may be associated with MGMT promoter methylation by transmethylation.

In this study, the T/N ratio correlated positively with the MGMT methylation rate in primary gliomas, but not in recurrent tumors. Various authors have assessed changes in MGMT promoter methylation status in paired initial and recurrent glioblastoma samples (7,8). Selective survival of tumor cells with high MGMT expression during alkylating agent therapy may lead to a change in MGMT status at recurrence (7,21,28).

In our previous study, we suggested that cell density in glioma tissue contributes to 11C-methionine uptake in stereotactic analysis (16). Furthermore, in another series of patients, including those with high grade gliomas, no statistically significant difference in T/N 11C-methionine uptake ratios were found between patients with and without MGMT methylation (29,30). Methionine uptake in gliomas is governed by changes in tracer influx across the blood-brain barrier (BBB) (31). In previous studies (16,29,30), the enhancing gliomas and methionine uptake were affected by BBB disruption and reflected the amino acid transport system and proliferation. Our present results analyzed non-enhancing gliomas and suggested that MGMT methylation tends to contribute markedly more to 11C-methionine uptake than to cell density in glioma tissue. However, the linear correlation of MGMT methylation and 11C-methionine uptake mainly arose from oligodendroglioma cases. An evaluation of the grading and histological types of gliomas and biopsy sites should be performed from this perspective.

This study was limited by its small sample size. Moreover, it remains unclear whether our results will be useful for prognosis and predicting response to therapy. Future large-scale studies are required to validate the proposed correlation between MGMT methylation and 11C-methionine uptake in a stereotactic manner.

To the best of our knowledge, no previous study has demonstrated stereotactic imaging of MGMT methylation status in gliomas using the noninvasive 11C-methionine PET imaging technique. When we investigated the correlation of the 11C-methionine T/N ratio in PET with quantitative MGMT promoter methylation for non-enhancing gliomas in a stereotactic image-based histological analysis, we found that the T/N ratio correlated positively with the MGMT methylation rate in primary gliomas. Our findings can contribute to understanding of the local characteristics of MGMT methylation status in primary and recurrent gliomas.

Acknowledgements

Not applicable.

Funding

This work was supported by the JSPS KAKENHI (grant nos. JP15K15534 and 16K20033).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

YO was responsible for the conception of the study and its design. YO and MN acquired the data. YO, MK, TS, DK, EY, YKo, MM and YKa, SN and TF analyzed and interpreted the data. YO drafted the manuscript. All authors critically revised the manuscript for important intellectual content and read and approved the final manuscript.

Ethics approval and consent to participate

The study was approved by the Ethical Committee of Osaka National Hospital (approval no. 94; IRB no. 0713). All patients provided written informed consent prior to their inclusion within the study.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

FLAIR

fluid-attenuated inversion recovery

MRI

magnetic resonance imaging

PET

positron-emission tomography

qMSP

quantitative methylation-specific PCR

SUV

standard uptake value

T/N ratio

tumor/normal tissue ratio

WHO

World Health Organization

References

1 

Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, Kros JM, Hainfellner JA, Mason W, Mariani L, et al: MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 352:997–1003. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Weller M, Tabatabai G, Kästner B, Felsberg J, Steinbach JP, Wick A, Schnell O, Hau P, Herrlinger U, Sabel MC, et al: MGMT Promoter methylation is a strong prognostic biomarker for benefit from dose-intensified temozolomide rechallenge in progressive glioblastoma: The DIRECTOR trial. Clin Cancer Res. 21:2057–2064. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Aum DJ, Kim DH, Beaumont TL, Leuthardt EC, Dunn GP and Kim AH: Molecular and cellular heterogeneity: The hallmark of glioblastoma. Neurosurg Focus. 37:E112014. View Article : Google Scholar : PubMed/NCBI

4 

Bonavia R, Inda MM, Cavenee WK and Furnari FB: Heterogeneity maintenance in glioblastoma: A social network. Cancer Res. 71:4055–4060. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Raja R, Sinha N, Saini J, Mahadevan A, Rao KN and Swaminathan A: Assessment of tissue heterogeneity using diffusion tensor and diffusion kurtosis imaging for grading gliomas. Neuroradiology. 58:1217–1231. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Parker NR, Hudson AL, Khong P, Parkinson JF, Dwight T, Ikin RJ, Zhu Y, Cheng ZJ, Vafaee F, Chen J, et al: Intratumoral heterogeneity identified at the epigenetic, genetic and transcriptional level in glioblastoma. Sci Rep. 6:224772016. View Article : Google Scholar : PubMed/NCBI

7 

Brandes AA, Franceschi E, Tosoni A, Bartolini S, Bacci A, Agati R, Ghimenton C, Turazzi S, Talacchi A, Skrap M, et al: O(6)-methylguanine DNA-methyltransferase methylation status can change between first surgery for newly diagnosed glioblastoma and second surgery for recurrence: Clinical implications. Neuro Oncol. 12:283–288. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Parkinson JF, Wheeler HR, Clarkson A, McKenzie CA, Biggs MT, Little NS, Cook RJ, Messina M, Robinson BG and McDonald KL: Variation of O(6)-methylguanine-DNA methyltransferase (MGMT) promoter methylation in serial samples in glioblastoma. J Neurooncol. 87:71–78. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Park CK, Kim JE, Kim JY, Song SW, Kim JW, Choi SH, Kim TM, Lee SH, Kim IH and Park SH: The changes in MGMT promoter methylation status in initial and recurrent glioblastomas. Transl Oncol. 5:393–397. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Andronesi OC, Kim GS, Gerstner E, Batchelor T, Tzika AA, Fantin VR, Vander Heiden MG and Sorensen AG: Detection of 2-hydroxyglutarate in IDH-mutated glioma patients by in vivo spectral-editing and 2D correlation magnetic resonance spectroscopy. Sci Transl Med. 4:116ra42012. View Article : Google Scholar : PubMed/NCBI

11 

Choi C, Ganji SK, DeBerardinis RJ, Hatanpaa KJ, Rakheja D, Kovacs Z, Yang XL, Mashimo T, Raisanen JM, Marin-Valencia I, et al: 2-hydroxyglutarate detection by magnetic resonance spectroscopy in IDH-mutated patients with gliomas. Nat Med. 18:624–629. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Okita Y, Nonaka M, Shofuda T, Kanematsu D, Yoshioka E, Kodama Y, Mano M, Nakajima S and Kanemura Y: (11)C-methinine uptake correlates with MGMT promoter methylation in nonenhancing gliomas. Clin Neurol Neurosurg. 125:212–216. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Pope WB, Prins RM, Albert Thomas M, Nagarajan R, Yen KE, Bittinger MA, Salamon N, Chou AP, Yong WH, Soto H, et al: Non-invasive detection of 2-hydroxyglutarate and other metabolites in IDH1 mutant glioma patients using magnetic resonance spectroscopy. J Neurooncol. 107:197–205. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Saito T, Maruyama T, Muragaki Y, Tanaka M, Nitta M, Shinoda J, Aki T, Iseki H, Kurisu K and Okada Y: 11C-methionine uptake correlates with combined 1p and 19q loss of heterozygosity in oligodendroglial tumors. AJNR Am J Neuroradiol. 34:85–91. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Shinozaki N, Uchino Y, Yoshikawa K, Matsutani T, Hasegawa A, Saeki N and Iwadate Y: Discrimination between low-grade oligodendrogliomas and diffuse astrocytoma with the aid of 11C-methionine positron emission tomography. J Neurosurg. 114:1640–1647. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Okita Y, Kinoshita M, Goto T, Kagawa N, Kishima H, Shimosegawa E, Hatazawa J, Hashimoto N and Yoshimine T: (11)C-methionine uptake correlates with tumor cell density rather than with microvessel density in glioma: A stereotactic image-histology comparison. Neuroimage. 49:2977–2982. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Esteller M, Hamilton SR, Burger PC, Baylin SB and Herman JG: Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. Cancer Res. 59:793–797. 1999.PubMed/NCBI

18 

Berger G, Maziere M, Knipper R, Prenant C and Comar D: Automated synthesis of 11C-labelled radiopharmaceuticals: Imipramine, chlorpromazine, nicotine and methionine. Int J Appl Radiat Isot. 30:393–399. 1979. View Article : Google Scholar : PubMed/NCBI

19 

Knudsen GM, Pettigrew KD, Patlak CS, Hertz MM and Paulson OB: Asymmetrical transport of amino acids across the blood-brain barrier in humans. J Cereb Blood Flow Metab. 10:698–706. 1990. View Article : Google Scholar : PubMed/NCBI

20 

Souba WW and Pacitti AJ: How amino acids get into cells: Mechanisms, models, menus, and mediators. JPEN J Parenter Enteral Nutr. 16:569–578. 1992. View Article : Google Scholar : PubMed/NCBI

21 

Christmann M, Nagel G, Horn S, Krahn U, Wiewrodt D, Sommer C and Kaina B: MGMT activity, promoter methylation and immunohistochemistry of pretreatment and recurrent malignant gliomas: A comparative study on astrocytoma and glioblastoma. Int J Cancer. 127:2106–2118. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Bergström M, Lundqvist H, Ericson K, Lilja A, Johnström P, Långström B, von Holst H, Eriksson L and Blomqvist G: Comparison of the accumulation kinetics of L-(methyl-11C)-methionine and D-(methyl-11C)-methionine in brain tumors studied with positron emission tomography. Acta Radiol. 28:225–229. 1987. View Article : Google Scholar : PubMed/NCBI

23 

Kato T, Shinoda J, Nakayama N, Miwa K, Okumura A, Yano H, Yoshimura S, Maruyama T, Muragaki Y and Iwama T: Metabolic assessment of gliomas using 11C-methionine, [18F] fluorodeoxyglucose and 11C-choline positron-emission tomography. AJNR Am J Neuroradiol. 29:1176–1182. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Ishiwata K, Ido T, Abe Y, Matsuzawa T and Iwata R: Tumor uptake studies of S-adenosyl-L-[methyl-11C]methionine and L-[methyl-11C]methionine. Int J Rad Appl Instrum B. 15:123–126. 1988. View Article : Google Scholar : PubMed/NCBI

25 

Baldessarini RJ and Kopin IJ: S-adenosylmethionine in brain and other tissues. J Neurochem. 13:769–777. 1966. View Article : Google Scholar : PubMed/NCBI

26 

Andreoli VM, Agnoli A and Fazio C: Transmethylations and the central nervous system. Springer-Verlag; Berlin; 1978, View Article : Google Scholar

27 

Ishiwata K, Vaalburg W, Elsinga PH, Paans AM and Woldring MG: Comparison of L-[1-11C]methionine and L-methyl-[11C]methionine for measuring in vivo protein synthesis rates with PET. J Nucl Med. 29:1419–1427. 1988.PubMed/NCBI

28 

Wiewrodt D, Nagel G, Dreimüller N, Hundsberger T, Perneczky A and Kaina B: MGMT in primary and recurrent human glioblastomas after radiation and chemotherapy and comparison with p53 status and clinical outcome. Int J Cancer. 122:1391–1399. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Choi H, Bang JI, Cheon GJ, Kim YH, Park CK, Park SH, Kang KW, Chung JK, Kim EE and Lee DS: 18F-fluorodeoxyglucose and11C-methionine positron emission tomography in relation to methyl-guanine methyltransferase promoter methylation in high-grade gliomas. Nucl Med Commun. 36:211–218. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Lopci E, Riva M, Olivari L, Raneri F, Soffietti R, Piccardo A, Bizzi A, Navarria P, Ascolese AM, Rudà R, et al: Prognostic value of molecular and imaging biomarkers in patients with supratentorial glioma. Eur J Nucl Med Mol Imaging. 44:1155–1164. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Roelcke U, Radü EW, von Ammon K, Hausmann O, Maguire RP and Leenders KL: Alteration of blood-brain barrier in human brain tumors: Comparison of [18F]fluorodeoxyglucose, [11C]methionine and rubidium-82 using PET. J Neurol Sci. 132:20–27. 1995. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 16 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Okita Y, Shofuda T, Kanematsu D, Yoshioka E, Kodama Y, Mano M, Kinoshita M, Nonaka M, Nakajima S, Fujinaka T, Fujinaka T, et al: Stereotactic image‑based histological analysis reveals a correlation between 11C‑methionine uptake and MGMT promoter methylation in non‑enhancing gliomas. Oncol Lett 16: 1924-1930, 2018.
APA
Okita, Y., Shofuda, T., Kanematsu, D., Yoshioka, E., Kodama, Y., Mano, M. ... Kanemura, Y. (2018). Stereotactic image‑based histological analysis reveals a correlation between 11C‑methionine uptake and MGMT promoter methylation in non‑enhancing gliomas. Oncology Letters, 16, 1924-1930. https://doi.org/10.3892/ol.2018.8866
MLA
Okita, Y., Shofuda, T., Kanematsu, D., Yoshioka, E., Kodama, Y., Mano, M., Kinoshita, M., Nonaka, M., Nakajima, S., Fujinaka, T., Kanemura, Y."Stereotactic image‑based histological analysis reveals a correlation between 11C‑methionine uptake and MGMT promoter methylation in non‑enhancing gliomas". Oncology Letters 16.2 (2018): 1924-1930.
Chicago
Okita, Y., Shofuda, T., Kanematsu, D., Yoshioka, E., Kodama, Y., Mano, M., Kinoshita, M., Nonaka, M., Nakajima, S., Fujinaka, T., Kanemura, Y."Stereotactic image‑based histological analysis reveals a correlation between 11C‑methionine uptake and MGMT promoter methylation in non‑enhancing gliomas". Oncology Letters 16, no. 2 (2018): 1924-1930. https://doi.org/10.3892/ol.2018.8866