Skip to main content

Advertisement

Log in

Quantitative mapping of individual voxels in the peritumoral region of IDH-wildtype glioblastoma to distinguish between tumor infiltration and edema

  • Clinical Study
  • Published:
Journal of Neuro-Oncology Aims and scope Submit manuscript

Abstract

Purpose

The peritumoral region (PTR) in glioblastoma (GBM) represents a combination of infiltrative tumor and vasogenic edema, which are indistinguishable on magnetic resonance imaging (MRI). We developed a radiomic signature by using imaging data from low grade glioma (LGG) (marker of tumor) and PTR of brain metastasis (BM) (marker of edema) and applied it on the GBM PTR to generate probabilistic maps.

Methods

270 features were extracted from T1-weighted, T2-weighted, and apparent diffusion coefficient maps in over 3.5 million voxels of LGG (36 segments) and BM (45 segments) scanned in a 1.5T MRI. A support vector machine classifier was used to develop the radiomics model from approximately 50% voxels (downsampled to 10%) and validated with the remaining. The model was applied to over 575,000 voxels of the PTR of 10 patients with GBM to generate a quantitative map using Platt scaling (infiltrative tumor vs. edema).

Results

The radiomics model had an accuracy of 0.92 and 0.79 in the training and test set, respectively (LGG vs. BM). When extrapolated on the GBM PTR, 9 of 10 patients had a higher percentage of voxels with a tumor-like signature over radiological recurrence areas. In 7 of 10 patients, the areas under curves (AUC) were > 0.50 confirming a positive correlation. Including all the voxels from the GBM patients, the infiltration signature had an AUC of 0.61 to predict recurrence.

Conclusion

A radiomic signature can demarcate areas of microscopic tumors from edema in the PTR of GBM, which correlates with areas of future recurrence.

Graphic abstract

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

Data availability

Data will be made available on request to the corresponding author following institutional ethics committee protocols.

Code availability

The radiomic feature extraction was performed using freely available Pyradiomics software (default set of features) (http://www.pyradiomics.io/pyradiomics.html). All standardization, model fitting, and assessment were performed using Scikit-Learn (https://scikit-learn.org/stable). The SVM classifier was used based on LIBSVM with a radial-basis-function kernel.

References

  1. Lambin P, Rios-Velazquez E, Leijenaar R et al (2012) Radiomics: Extracting more information from medical images using advanced feature analysis. Eur J Cancer 48:441–446. https://doi.org/10.1016/j.ejca.2011.11.036

    Article  PubMed  PubMed Central  Google Scholar 

  2. Villanueva-Meyer JE, Mabray MC, Cha S (2017) Current clinical brain tumor imaging. Neurosurgery 81:397–415. https://doi.org/10.1093/neuros/nyx103

    Article  PubMed  Google Scholar 

  3. Chaddad A, Kucharczyk MJ, Daniel P et al (2019) Radiomics in glioblastoma: current status and challenges facing clinical implementation. Front Oncol. https://doi.org/10.3389/fonc.2019.00374

    Article  PubMed  Google Scholar 

  4. Yamahara T, Numa Y, Oishi T et al (2010) Morphological and flow cytometric analysis of cell infiltration in glioblastoma: a comparison of autopsy brain and neuroimaging. Brain Tumor Pathol 27:81–87. https://doi.org/10.1007/s10014-010-0275-7

    Article  PubMed  Google Scholar 

  5. Barajas RF, Phillips JJ, Parvataneni R et al (2012) Regional variation in histopathologic features of tumor specimens from treatment-naive glioblastoma correlates with anatomic and physiologic MR Imaging. Neuro Oncol 14:942–954. https://doi.org/10.1093/neuonc/nos128

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Eidel O, Burth S, Neumann J-O et al (2017) Tumor infiltration in enhancing and non-enhancing parts of glioblastoma: a correlation with histopathology. PLoS ONE. https://doi.org/10.1371/journal.pone.0169292

    Article  PubMed  Google Scholar 

  7. Duron L, Balvay D, Perre SV et al (2019) Gray-level discretization impacts reproducible MRI radiomics texture features. PLoS ONE. https://doi.org/10.1371/journal.pone.0213459

    Article  PubMed  Google Scholar 

  8. Carré A, Klausner G, Edjlali M et al (2020) Standardization of brain MR images across machines and protocols: bridging the gap for MRI-based radiomics. Sci Rep. https://doi.org/10.1038/s41598-020-69298-z

    Article  PubMed  Google Scholar 

  9. McDonald MW, Shu H-KG, Curran WJ, Crocker IR (2011) Pattern of failure after limited margin radiotherapy and temozolomide for glioblastoma. Int J Radiat Oncol Biol Phys 79:130–136. https://doi.org/10.1016/j.ijrobp.2009.10.048

    Article  CAS  PubMed  Google Scholar 

  10. Petrecca K, Guiot M-C, Panet-Raymond V, Souhami L (2013) Failure pattern following complete resection plus radiotherapy and temozolomide is at the resection margin in patients with glioblastoma. J Neurooncol 111:19–23. https://doi.org/10.1007/s11060-012-0983-4

    Article  PubMed  Google Scholar 

  11. Lemée J-M, Clavreul A, Menei P (2015) Intratumoral heterogeneity in glioblastoma: don’t forget the peritumoral brain zone. Neuro Oncol 17:1322–1332. https://doi.org/10.1093/neuonc/nov119

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Glas M, Rath BH, Simon M et al (2010) Residual tumor cells are unique cellular targets in glioblastoma. Ann Neurol 68:264–269. https://doi.org/10.1002/ana.22036

    Article  PubMed  PubMed Central  Google Scholar 

  13. Ruiz-Ontañon P, Orgaz JL, Aldaz B et al (2013) Cellular plasticity confers migratory and invasive advantages to a population of glioblastoma-initiating cells that infiltrate peritumoral tissue. Stem Cells 31:1075–1085. https://doi.org/10.1002/stem.1349

    Article  CAS  PubMed  Google Scholar 

  14. Price SJ, Jena R, Burnet NG et al (2006) Improved delineation of glioma margins and regions of infiltration with the use of diffusion tensor imaging: an image-guided biopsy study. AJNR Am J Neuroradiol 27:1969–1974

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Yan J-L, Li C, Boonzaier NR et al (2019) Multimodal MRI characteristics of the glioblastoma infiltration beyond contrast enhancement. Ther Adv Neurol Disord 12:1756286419844664. https://doi.org/10.1177/1756286419844664

    Article  PubMed  PubMed Central  Google Scholar 

  16. Blystad I, Warntjes JBM, Smedby Ö et al (2020) Quantitative MRI using relaxometry in malignant gliomas detects contrast enhancement in peritumoral oedema. Sci Rep 10:17986. https://doi.org/10.1038/s41598-020-75105-6

    Article  CAS  PubMed  Google Scholar 

  17. Akbari H, Macyszyn L, Da X et al (2016) Imaging surrogates of infiltration obtained via multiparametric imaging pattern analysis predict subsequent location of recurrence of glioblastoma. Neurosurgery 78:572–580. https://doi.org/10.1227/NEU.0000000000001202

    Article  PubMed  Google Scholar 

  18. Rathore S, Akbari H, Doshi J, et al (2018) Radiomic signature of infiltration in peritumoral edema predicts subsequent recurrence in glioblastoma: implications for personalized radiotherapy planning. JMI 5:021219. https://doi.org/10.1117/1.JMI.5.2.021219

    Article  PubMed  Google Scholar 

  19. Hu LS, Ning S, Eschbacher JM et al (2015) Multi-parametric MRI and texture analysis to visualize spatial histologic heterogeneity and tumor extent in glioblastoma. PLoS ONE 10:e0141506. https://doi.org/10.1371/journal.pone.0141506

    Article  CAS  PubMed  Google Scholar 

  20. Yan J-L, Li C, van der Hoorn A et al (2020) A neural network approach to identify the peritumoral invasive areas in glioblastoma patients by using MR radiomics. Sci Rep 10:9748. https://doi.org/10.1038/s41598-020-66691-6

    Article  CAS  PubMed  Google Scholar 

  21. Stewart J, Sahgal A, Lee Y et al (2020) Quantitating inter-fraction target dynamics during concurrent chemoradiation for glioblastoma: a prospective serial imaging study. Int J Radiat Oncol Biol Phys. https://doi.org/10.1016/j.ijrobp.2020.10.002

    Article  PubMed  Google Scholar 

  22. Jackson C, Choi J, Khalafallah AM et al (2020) A systematic review and meta-analysis of supratotal versus gross total resection for glioblastoma. J Neurooncol 148:419–431. https://doi.org/10.1007/s11060-020-03556-y

    Article  PubMed  Google Scholar 

  23. Certo F, Altieri R, Maione M et al (2020) FLAIRectomy in supramarginal resection of glioblastoma correlates with clinical outcome and survival analysis: a prospective, single institution, Case Series. Oper Neurosurg (Hagerstown). https://doi.org/10.1093/ons/opaa293

    Article  Google Scholar 

  24. Tseng C-L, Stewart J, Whitfield G et al (2020) Glioma consensus contouring recommendations from a MR-Linac International Consortium Research Group and evaluation of a CT-MRI and MRI-only workflow. J Neurooncol. https://doi.org/10.1007/s11060-020-03605-6

    Article  PubMed  PubMed Central  Google Scholar 

  25. Kumar N, Kumar R, Sharma SC et al (2020) Impact of volume of irradiation on survival and quality of life in glioblastoma: a prospective, phase 2, randomized comparison of RTOG and MDACC protocols. Neurooncol Pract 7:86–93. https://doi.org/10.1093/nop/npz024

    Article  PubMed  Google Scholar 

  26. Azoulay M, Chang SD, Gibbs IC et al (2020) A phase I/II trial of 5-fraction stereotactic radiosurgery with 5-mm margins with concurrent temozolomide in newly diagnosed glioblastoma: primary outcomes. Neuro Oncol 22:1182–1189. https://doi.org/10.1093/neuonc/noaa019

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank the patients and their caregivers involved in the study. Our sincere gratitude to the Terry Fox Foundation Program Project Grant from the Hecht Foundation for the funding support associated with the study.

Funding

Terry Fox Foundation Program Project Grant from the Hecht Foundation (1083) awarded to Gregory J. Czarnota. The funding bodies had no influence on the study design, data collection, analysis, interpretation of data, or the manuscript's writing.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: AD, BG, AS, GJC; Methodology: All authors; Formal Analysis and investigation: All authors; Writing-original draft preparation: AD, BG, AS, GJC; Writing-review and editing: All authors; Project administration and supervision: AS, GJC; Funding acquisition: GJC. All the authors are in agreement and accountable for all the aspects of the work.

Corresponding author

Correspondence to Gregory J. Czarnota.

Ethics declarations

Conflict of interest

Archya Dasgupta: None. Benjamin Geraghty: None. Pejman Maralani: None. Nauman Malik: None. Michael Sandhu: None. Jay Detsky: None. Chia-Lin Tseng: Travel accommodations/expenses & honoraria for past educational seminars by Elekta, belongs to the Elekta MR-Linac Research Consortium, and advisor/consultant with Sanofi. Hany Soliman: None. Sten Myrehaug: Travel accommodations/expenses from Elekta AB. Research support from Novartis/AAA. Zain Husain: Travel accommodations/expenses from Elekta. James Perry: None. Angus Lau: None. Arjun Sahgal: Advisor/consultant with AbbVie, Merck, Roche, Varian (Medical Advisory Group), Elekta (Gamma Knife Icon), BrainLAB, and VieCure (Medical Advisory Board). Board Member: International Stereotactic Radiosurgery Society (ISRS). Past educational seminars with Elekta AB, Accuray Inc., Varian (CNS Teaching Faculty), BrainLAB, Medtronic Kyphon. Research grant with Elekta AB. Travel accommodations/expenses by Elekta, Varian, BrainLAB. Elekta MR Linac Research Consortium, Elekta Spine, Oligometastases and Linac Based SRS Consortia. Gregory J. Czarnota: Funding received from the Terry Fox Foundation Program Project Grant.

Ethical approval

The study was approved by the Research Ethics Board of Sunnybrook Health Sciences Centre (Protocol Number: 034-2020).

Consent to participate

Consent was waived for the retrospective study.

Consent for publication

Not applicable (anonymized data, imaging study).

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file 1 (DOCX 29 kb)

11060_2021_3762_MOESM2_ESM.pptx

Supplementary file 2—Supplementary Figure 1 shows the receiver operating characteristics curve in the test set of low grade glioma and peritumoral region of brain metastases for voxel-based classification (PPTX 112 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dasgupta, A., Geraghty, B., Maralani, P.J. et al. Quantitative mapping of individual voxels in the peritumoral region of IDH-wildtype glioblastoma to distinguish between tumor infiltration and edema. J Neurooncol 153, 251–261 (2021). https://doi.org/10.1007/s11060-021-03762-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11060-021-03762-2

Keywords

Navigation